Language selection

Search

Patent 2703257 Summary

Third-party information liability

Some of the information on this Web page has been provided by external sources. The Government of Canada is not responsible for the accuracy, reliability or currency of the information supplied by external sources. Users wishing to rely upon this information should consult directly with the source of the information. Content provided by external sources is not subject to official languages, privacy and accessibility requirements.

Claims and Abstract availability

Any discrepancies in the text and image of the Claims and Abstract are due to differing posting times. Text of the Claims and Abstract are posted:

  • At the time the application is open to public inspection;
  • At the time of issue of the patent (grant).
(12) Patent: (11) CA 2703257
(54) English Title: BENZOMORPHOLINE DERIVATIVES AND METHODS OF USE
(54) French Title: DERIVES DE BENZOMORPHOLINE ET PROCEDES D'UTILISATION
Status: Deemed expired
Bibliographic Data
(51) International Patent Classification (IPC):
  • C07D 413/14 (2006.01)
  • A61K 31/536 (2006.01)
  • A61P 35/00 (2006.01)
(72) Inventors :
  • HARMANGE, JEAN-CHRISTOPHE (United States of America)
  • MARTIN, MATTHEW W. (United States of America)
  • TEFFERA, YOHANNES (United States of America)
  • SUBRAMANIAN, RAJU (United States of America)
  • WHITE, RYAN (United States of America)
  • ZANON, ROGER (United States of America)
  • LARROW, JAY (United States of America)
  • PAYACK, JOSEPH F. (United States of America)
  • DILMEGHANI SERAN, MINA (United States of America)
(73) Owners :
  • AMGEN INC. (United States of America)
(71) Applicants :
  • AMGEN INC. (United States of America)
(74) Agent: GOWLING WLG (CANADA) LLP
(74) Associate agent:
(45) Issued: 2013-02-19
(86) PCT Filing Date: 2008-10-28
(87) Open to Public Inspection: 2009-05-07
Examination requested: 2010-04-20
Availability of licence: N/A
(25) Language of filing: English

Patent Cooperation Treaty (PCT): Yes
(86) PCT Filing Number: PCT/US2008/012224
(87) International Publication Number: WO2009/058267
(85) National Entry: 2010-04-20

(30) Application Priority Data:
Application No. Country/Territory Date
61/000,948 United States of America 2007-10-29

Abstracts

English Abstract




Selected benzomorpholine compounds are effective for prophylaxis and treatment
of diseases, such as VEGF mediated
diseases. The invention encompasses novel compounds, analogs, prodrugs and
pharmaceutically acceptable salts thereof,
pharmaceutical compositions and methods for prophylaxis and treatment of
diseases and other maladies or conditions involving,
cancer and the like. The subject invention also relates to processes for
making such compounds as well as to intermediates useful in
such processes.


French Abstract

La présente invention concerne des composés de benzomorpholine efficaces dans la prophylaxie et le traitement de maladies, telles que des maladies induites par le VEGF. L'invention concerne également de nouveaux composés, analogues et promédicaments, ainsi que des sels pharmaceutiquement acceptables de ceux-ci, des compositions pharmaceutiques et des procédés de prophylaxie et de traitement de maladies et d'autres maladies ou états impliquant le cancer et analogue. La présente invention porte en outre sur des procédés de réalisation de tels composés, ainsi que sur des intermédiaires utiles dans de tels procédés.

Claims

Note: Claims are shown in the official language in which they were submitted.



CLAIMS:
1. A compound selected from

Image
and pharmaceutically acceptable salts thereof.

2. A compound of claim 1 wherein said compound is the hydrochloride salt
of the compound.

3. A compound of claim 1 wherein said compound is selected from
57


Image
and pharmaceutically acceptable salts thereof.

4. A pharmaceutical composition comprising a compound of claim 1
together with a pharmaceutically acceptable vehicle adjuvant or diluent.

5. A use of an effective amount of a compound of claim 1, for treating
tumors in a patient in need of such treatment.

6. A use of an effective amount of a compound of claim 1, for the
preparation of a medicament for treating tumors in a patient in need of such
treatment.
7. The use of claim 5 or 6 wherein angiogenesis is inhibited.

8. The use of claim 5 or 6 wherein tumor growth is inhibited.
9. The use of claim 5 or 6 wherein tumor size is reduced.

10. The use of claim 5 or 6 wherein metastasis is inhibited.
58


11. The use of claim 5 or 6 wherein the compound of claim 1 is used in
combination with at least one additional therapeutic agent.

12. A compound of claim 1 and pharmaceutically acceptable salts thereof
wherein said compound is 7-((6,7-bis(methyloxy)-4-quinolinyl)oxy)-N-(5-methyl-
3-
isoxazolyl)-2,3-dihydro-4H-1,4-benzoxazine-4-carboxamide.

59

Description

Note: Descriptions are shown in the official language in which they were submitted.



CA 02703257 2010-04-20
WO 2009/058267 PCT/US2008/012224
BENZOMORPHOLINE DERIVATIVES AND METHODS OF USE
FIELD OF THE INVENTION
This invention is in the field of pharmaceutical agents and specifically
relates to
compounds, compositions, uses and methods for treating cancer.
BACKGROUND OF THE INVENTION
Protein kinases represent a large family of proteins which play a central role
in the
regulation of a wide variety of cellular processes, maintaining control over
cellular function. A
partial list of such kinases includes abl, Akt, bcr-abl, Blk, Brk, Btk, c-kit,
c-Met, c-src, c-fins,
CDKI, CDK2, CDK3, CDK4, CDK5, CDK6, CDK7, CDK8, CDK9, CDK10, cRafl, CSF1R,
CSK, EGFR, ErbB2, ErbB3, ErbB4, Erk, Fak, fes, FGFRI, FGFR2, FGFR3, FGFR4,
FGFR5,
Fgr, fit-1, Fps, Frk, Fyn, Hck, IGF-IR, INS-R, Jak, KDR, Lck, Lyn, MEK, p38,
PDGFR, PIK,
PKC, PYK2, ros, tie, tie2, TRK, Yes, and Zap70. Inhibition of such kinases has
become an
important therapeutic target.
Certain diseases are known to be associated with deregulated angiogenesis, for
example
ocular neovascularisation, such as retinopathies (including diabetic
retinopathy), age-related
macular degeneration, psoriasis, hemangioblastoma, hemangioma,
arteriosclerosis,
inflammatory disease, such as a rheumatoid or rheumatic inflammatory disease,
especially
arthritis (including rheumatoid arthritis), or other chronic inflammatory
disorders, such as
chronic asthma, arterial or post-transplantational atherosclerosis,
endometriosis, and neoplastic
diseases, for example so-called solid tumors and liquid tumors (such as
leukemias).
At the center of the network regulating the growth and differentiation of the
vascular
system and its components, both during embryonic development and normal
growth, and in a
wide number of pathological anomalies and diseases, lies the angiogenic factor
known as
Vascular Endothelial Growth Factor"(VEGF; originally termed 'Vascular
Permeability Factor",
VPF), along with its cellular receptors (see G. Breier et al., Trends in Cell
Biology, 6:454-456
(1996)).
VEGF is a dimeric, disulfide-linked 46-kDa glycoprotein related to "Platelet-
Derived
Growth Factor" (PDGF); it is produced by normal cell lines and tumor cell
lines; is an
endothelial cell-specific mitogen; shows angiogenic activity in in vivo test
systems (e.g. rabbit
cornea); is chemotactic for endothelial cells and monocytes; and induces
plasminogen
activators in endothelial cells, which are involved in the proteolytic
degradation of
extracellular matrix during the formation of capillaries. A number of isoforms
of VEGF are
known, which show comparable biological activity, but differ in the type of
cells that secrete


CA 02703257 2010-04-20
WO 2009/058267 PCT/US2008/012224
them and in their heparin-binding capacity. In addition, there are other
members of the VEGF
family, such as "Placenta Growth Factor"(PIGF) and VEGF-C.
VEGF receptors (VEGFR) are transmembranous receptor tyrosine kinases. They are
characterized by an extracellular domain with seven immunoglobulin-like
domains and an
intracellular tyrosine kinase domain. Various types of VEGF receptor are
known, e.g.
VEGFR-1 (also known as flt-1), VEGFR-2 (also known as KDR), and VEGFR-3.
A large number of human tumors, especially gliomas and carcinomas, express
high
levels of VEGF and its receptors. This has led to the hypothesis that the VEGF
released by
tumor cells stimulates the growth of blood capillaries and the proliferation
of tumor
endothelium in a paracrine manner and through the improved blood supply,
accelerate tumor
growth. Increased VEGF expression could explain the occurrence of cerebral
edema in
patients with glioma. Direct evidence of the role of VEGF as a tumor
angiogenesis factor in
vivo is shown in studies in which VEGF expression or VEGF activity was
inhibited. This was
achieved with anti-VEGF antibodies, with dominant-negative VEGFR-2 mutants
which
inhibited signal transduction, and with antisense-VEGF RNA techniques. All
approaches led
to a reduction in the growth of glioma cell lines or other tumor cell lines in
vivo as a result of
inhibited tumor angiogenesis.
Angiogenesis is regarded as an absolute prerequisite for tumors which grow
beyond a
diameter of about 1-2 mm; up to this limit, oxygen and nutrients may be
supplied to the tumor
cells by diffusion. Every tumor, regardless of its origin and its cause, is
thus dependent on
angiogenesis for its growth after it has reached a certain size.
Three principal mechanisms play an important part in the activity of
angiogenesis
inhibitors against tumors: 1) Inhibition of the growth of vessels, especially
capillaries, into
avascular resting tumors, with the result that there is no net tumor growth
owing to the balance
that is achieved between cell death and proliferation; 2) Prevention of the
migration of tumor
cells owing to the absence of blood flow to and from tumors; and 3) Inhibition
of endothelial
cell proliferation, thus avoiding the paracrine growth-stimulating effect
exerted on the
surrounding tissue by the endothelial cells which normally line the vessels.
See R. Connell
and J. Beebe, Exp. Opin. Ther. Patents, 11:77-114 (2001).
VEGF's are unique in that they are the only angiogenic growth factors known to
contribute to vascular hyperpermeability and the formation of edema. Indeed,
vascular
hyperpermeability and edema that is associated with the expression or
administration of many
other growth factors appears to be mediated via VEGF production.

2


CA 02703257 2012-03-13

WO 2009/058267 PCT/US2008/012224
Inflammatory cytokines stimulate VEGF production. Hypoxia results in a marked
upregulation of VEGF in numerous tissues, hence situations involving infarct,
occlusion,
ischemia, anemia, or circulatory impairment typically invoke VEGF/VPF-mediated
responses.
Vascular hyperpermeability, associated edema, altered transendothelial
exchange and
macromolecular extravasation, which is often accompanied by diapedesis, can
result in
excessive matrix deposition, aberrant stromal proliferation, fibrosis, etc.
Hence, VEGF-
mediated hyperpermeability can significantly contribute to disorders with
these etiologic
features. As such, regulators of angiogenesis have become an important
therapeutic target.
Angiogenesis, the process of sprouting new blood vessels from existing
vasculature and
arter iogenesis, the remodeling of small vessels into larger conduit vessels
are both
physiologically important aspects of vascular growth in adult tissues. These
processes of
vascular growth are required for beneficial processes such as tissue repair,
wound healing,
recovery from tissue ischemia and menstrual cycling. They are also required
for the
development of pathological conditions such as the growth of neoplasias,
diabetic retinopathy,
rheumatoid arthritis, psoriasis, certain forms of macular degeneration, and
certain
inflammatory pathologies. The inhibition of vascular growth in these contexts
has also shown
beneficial effects in preclinical animal models. For example, inhibition of
angiogenesis by
blocking vascular endothelial growth factor or its receptor has resulted in
inhibition of tumor
growth and in retinopathy. Also, the development of pathological pannus tissue
in rheumatoid
arthritis involves angiogenesis and might be blocked by inhibitors of
angiogenesis.
The ability to stimulate vascular growth has potential utility for treatment
of ischemia-
induced pathologies such as myocardial infarction, coronary artery disease,
peripheral vascular
disease, and stroke. The sprouting of new vessels and/or the expansion of
small vessels in
ischemic tissues prevents ischemic tissue death and induces tissue repair.
Certain diseases are
known to be associated with deregulated angiogenesis, for example ocular
neovascularization,
such as retinopathies (including diabetic retinopathy), age-related macular
degeneration,
psoriasis, hemangioblastoma, hemangioma, arteriosclerosis, inflammatory
disease, such as a
rheumatoid or rheumatic inflammatory disease, especially arthritis (including
rheumatoid
arthritis), or other chronic inflammatory disorders, such as chronic asthma,
arterial or post-
transplantational atherosclerosis, endometriosis, and neoplastic diseases, for
example so-called
solid tumors and liquid tumors (such as leukemias).
WO 05/070891 describes certain benzomorpholine derivatives that are useful as
VEGF inhibitors, including the following reference compound 1:

3


CA 02703257 2010-04-20
WO 2009/058267 PCT/US2008/012224
H
IN N
\
O
N

O
O N

WO 05/070891 Ex. 781
Ref. Cmpd I

The benzomorpholine compounds of the current invention possess unexpected
advantages when compared to the closest compound in the prior art, reference
compound 1.
DESCRIPTION OF THE INVENTION
A class of compounds useful in treating cancer and angiogenesis is defined by
the following
compound A
H
O N N

O
N

O O
O \

N Compound A
active metabolites, salts and solvates thereof.
The present invention also relates to pharmaceutical compositions containing
the
above-described compounds, together with a pharmaceutically acceptable vehicle
or carrier.
The invention also relates to a method of treating cancer in a subject using
the above
compounds either alone or in combination with another therapeutic agent.
The invention also relates to a method of inhibiting angiogenesis in a subject
using the
above compounds either alone or in combination with another therapeutic agent.
The invention also relates to a method of treating tumors in a subject using
the above
compounds either alone or in combination with another therapeutic agent.

4


CA 02703257 2010-04-20
WO 2009/058267 PCT/US2008/012224

The invention also relates to a method of inhibiting tumor growth in a subject
using the
above compounds either alone or in combination with another therapeutic agent.
The invention also relates to a method of reducing tumor size in a subject
using the
above compounds either alone or in combination with another therapeutic agent.
The invention also relates to a method of inhibiting metastasis of tumors in a
subject
using the above compounds either alone or in combination with another
therapeutic agent.
The invention also relates to a method of treating VEGF-mediated disorders in
a
subject using the above compounds either alone or in combination with another
therapeutic
agent.
The invention further relates to a process for preparing compound A,
salts, and solvates thereof comprising the step of contacting a compound of
the following
formula I
H
N
O O
O

O N~ I
with a compound of the following formula II
O NCO
RX

O N
H II
wherein RX is optionally substituted aryl or heteroaryl;

in the presence of
(1) a polar solvent ; and
(2) a base

Suitable polar solvents include, but are not limited to esters, such as alkyl
acetates (e.g.,
methyl acteate, ethyl acetate, isopropyl acetate and the like), amides (such
as dimethyl
formamide, N-methyl pyrrolidinone, dimethyl acetamide and the like),
chlorinated
hydrocarbons (such as chlorinated benzene, methylene chloride, dichloro ethane
and the like),
5


CA 02703257 2010-04-20
WO 2009/058267 PCT/US2008/012224
ethers (such as methyl-t-butyl ether, tetrahydrofuran and the like), pyridine
and n-methyl
morpholine, or any combination thereof. Preferred polar solvents include ethyl
acetate,
isopropyl acetate and N-methyl pyrrolidinone ("NMP").
Suitable bases include, but are not limited to metal hydroxides (such as
lithium
hydroxide, sodium hydroxide, potassium hydroxide, magnesium hydroxide and the
like),
organic tertiary amines (such as triethyl amine, diisopropyl amine and the
like), metal
alkoxides (such as potassium or sodium methoxide, ethoxide, t-butoxide and the
like), metal
carbonates (such as sodium or potassium carbonate and the like), bicarbonates
(such as sodium
or potassium bicarbonate and the like), lithium amides (such as lithium
diisopropylamide and
the like), lithium alkyls (such as butyl lithium and the like), pyridine and N-
methyl
morpholine, or any combination thereof. Preferred bases include sodium
hydroxide, potassium
hydroxide, sodium t-butoxide, potassium t-butoxide, sodium carbonate and
potassium
carbonate.

The present invention further relates to a process wherein the compound of
formula 11
is
NCO
O

O N
H
The present invention further relates to a process wherein the compound of
formula II

is prepared by contacting

N-'O
H2N

with a compound of the following formula III
Ou
0 Halogen III
in the presence of
(1) a polar solvent ; and

6


CA 02703257 2010-04-20
WO 2009/058267 PCT/US2008/012224
(2) a base.

The present invention further relates to a process wherein the compound of
formula III
is

O
O CI
The present invention further relates to a process wherein the compound of
formula I is
prepared by contacting a compound of formula IV
H
N
HO O IV

with a compound of formula V
C1
0 N V

in the presence of N-methyl pyrrolidinone and potassium t-butoxide.

The present invention further relates to a process wherein the compound of
formula IV
is prepared by contacting a compound of formula VI
H
N O
Ho Co::r
VI
with BH3, MeOH and HCl .

The present invention further relates to a process wherein the compound of
formula VI
is prepared by contacting a compound of formula VII
NH2
HO OH VII

7


CA 02703257 2010-04-20
WO 2009/058267 PCT/US2008/012224
with a compound of formula VIII
O
CI '-Ilk~ CI
VIII
in the presence of potassium carbonate and toluene.

The invention further relates to a compound made by any of the above-described
processes.

We have discovered that compound A and salts thereof possess striking
unexpected
advantages when compared to the structurally closest compound found in the
prior art, i.e.,
reference compound 1. Specifically, compounds of the present invention exhibit
remarkably
improved in vivo characteristics when compared to reference compound 1.
Compound A and reference compound I both potently inhibit VEGF induced
proliferation in in vitro cell-based assays as shown below in Table 1 (this
assay is described in
WO 05/070891).
TABLE 1
Compound HUVEC IC50 (nM)
Reference Compound 1 7
Compound A 2

Tables 2 through 6 below describe the results obtained in the rat corneal in
vivo model
of angiogenesis for (1) reference compound (free base) 1, (2) Compound A (free
base), and (3)
the hydrochloride salt of Compound A. A VEGF-induced rat corneal angiogenesis
assay was
performed using the following procedure:
In Life Aspects: Female CD rats weighing approximately 250 grams were
randomized into one of six treatment groups. On the day of surgery the rats
were temporarily anesthetized using isoflurane. A vertical incision was made
on the midline of the cornea and a pocket was created to separate the
connective tissue layers of the stroma,. The distance between the apex of the
pocket and the limbus was approximately 1.8 mm. A presoaked nylon filter
disk was inserted to the apex of the pocket (in the case of the control
treatment
group, the disks were soaked in a buffer containing bovine serum albumin; in

8


CA 02703257 2010-04-20
WO 2009/058267 PCT/US2008/012224
the remaining treatment groups, the disks were soaked in buffer containing
bovine serum albumin plus human VEGF to induce angiogenesis). The
different treatment groups were then dosed daily with either vehicle (Ora
Plus"', a comer'cially available vehicle) or a designated amount of test
compound formulated in vehicle.

Study Termination and Analysis: After seven days, the rats were euthanized
and the implanted corneas photographed using a Nikon SV-3 Ophthalmic Slit
Lamp. Numerical data were generated from the digital images using the
Metamorph image analysis system (Universal Imaging). Three endpoints were
analyzed on each corneal image: (1) disk placement distance from the limbus,
(2) number of vessels intersecting a perpendicular line at the midpoint of the
disk placement distance, and (3) blood vessel area, as determined by
thresholding and automated pixel counting. Only the number of blood vessels
is shown in the tables as this endpoint correlated well with the blood vessel

area.
Statistical Analysis: Results were analyzed with the StatView statistical
program using one-way ANOVA, followed by Fisher's least significant
difference test. Data are presented as mean SE and P<0.05 was considered
significant.
AUC and ED50 values were calculated using methods well known in the art.
9


CA 02703257 2010-04-20
WO 2009/058267 PCT/US2008/012224
TABLE 2
Reference Compound 1 (Free Base)
Treatment Group # of Average # Standard AUC
Rats in of Vessels Error (ng*hr/
Group / ml)
Control disc + Vehicle 7 5.7 3.0
/NA
VEGF disc + Vehicle 7 19.7 4.7
/NA
VEGF disc + 0.01 mg/kg 7 22.0 3.9 0
Ref. Cmpd 1 /NS
VEGF disc + 0.03 mg/kg 7 25.4 3.6 22.5
Ref. Cmpd 1 /NS
VEGF disc + 0.1 mg/kg 7 19.3 2.6 68.5
Ref. Cmpd I /NS
VEGF disc + 0.3 mg/kg 8 15.9 4.0 204.6
Ref. Cmpd I /NS
ED50> 0.3 mg/kg (collected data did not allow for calculation of the ED50.
Fifty percent
reduction in the number of vessels formed did not occur at the highest dose
tested).
NS, not significant; NA, not applicable.



CA 02703257 2010-04-20
WO 2009/058267 PCT/US2008/012224
TABLE 3
Compound A (Free Base)
Treatment Group # of Average Standard AUC
Rats in # of Error (ng*hr/
ml
Group Vessels / p
Control disc + Vehicle 7 8.6 3.8
/NA

VEGF disc + Vehicle. 5 37.2 4.3
/NA
VEGF disc + 0.03 mg/kg 8 24.5 4.4 108.9
Cmpd A / =0.0477

VEGF disc + 0.1 mg/kg 8 24.9 5.6 203
Cmpd A /=0.0543

VEGF disc + 0.3 mg/kg 7 5.4 2.1 729.5
Cmpd A / <0.0001

VEGF disc + 1.0 mg/kg 6 2.3 2.0 2368.9
Cmpd A / <0.0001

ED50 = 0.16 mg/kg
AUC at ED50=400 ng*hr/ml
NS, not significant; NA, not applicable.

11


CA 02703257 2010-04-20
WO 2009/058267 PCT/US2008/012224
TABLE 4
Compound A (HCl Salt)
Run #1
Treatment Group # of Average Standard AUC
Rats in # of Error (ng*hr/
Group Vessels / ml)
Control disc + Vehicle 8 13.1 5.9
/NA
VEGF disc + Vehicle 8 26.8 4.6
/NA
VEGF disc + 0.03 mg/kg 6 15.3 4.7 161.7
Cmpd A /NS
VEGF disc + 0.1 mg/kg 8 9.3 3.9 459.9
Cmpd A / =0.0050

VEGF disc + 0.3 mg/kg 8 4.3 2.4 1187.9
Cmpd A / =0.0004
VEGF disc + 1.0 mg/kg 8 0.4 0.4 3638.2
Cmpd A / <0.0001

ED50 = 0.04 mg/kg
AUC at ED50=200 ng*hr/ml
NS, not significant; NA, not applicable.

12


CA 02703257 2010-04-20
WO 2009/058267 PCT/US2008/012224
TABLE 5
Compound A (HCl Salt)
Run #2
Treatment Group # of Average Standard AUC
Rats in # of Error (ng*hr/
Group Vessels / p ml)
Control disc +- Vehicle 8 5.4 2.6
/NA
VEGF disc + Vehicle 8 28.3 3.3
/NA
VEGF disc + 0.01 mg/kg 7 28.9 4.4 95.8
Cmpd A /NS
VEGF disc + 0.03 mg/kg 8 25.5 4.7 272.1
Cmpd A /NS
VEGF disc + 0.1 mg/kg 7 18.0 5.5 954.3
Cmpd A /NS

VEGF disc + 0.3 mg/kg 8 7.3 3.8 3899.5
Cmpd A / =0.0006

ED50 = 0.15 mg/kg
.;o=2000 ng*hr/ml
AUC at ED,
NS, not significant; NA, not applicable.

13


CA 02703257 2010-04-20
WO 2009/058267 PCT/US2008/012224
TABLE 6
Compound A (HCl Salt)
Run #3
Treatment Group # of Average Standard AUC
Rats in # of Error (ng*hr/
Group Vessels / ml
Control disc + Vehicle 7 2.7 1.0
/NA
VEGF disc + Vehicle 7 24.7 2.7
/NA
VEGF disc + 0.01 mg/kg 8 28.4 3.6 21.8
Cmpd A /NS
VEGF disc + 0.03 mg/kg 8 32.0 3.3 80.2
Cmpd A /NS
VEGF disc + 0.1 mg/kg 8 15.4 4.1 321.8
Cmpd A / =0.0307
VEGF disc + 0.3 mg/kg 8 4.8 1.6 677.4
Cmpd A /<0.0001

ED50= 0.14 mg/kg
AUC at ED50=338 ng*hr/ml
NS, not significant; NA, not applicable.

Two major points can be seen from a review of Tables 2 and 3:
(1) Compound A (free base) has a considerably lower ED50 than reference
compound 1
(free base), 0.16 mpk vs > 0.3 mpk; and
(2) at the same doses Compound A (free base) has consistently higher exposure
across
tested doses than that observed for reference compound I (free base). This
observation
correlates with the superior pharmacokinetic profile of compound A in rat
compared to the
reference compound I as indicated in Table 7.
Tables 4-6 illustrate the pharmacological activity of compound A in the rat
corneal
angiogenesis assay when dosed as an HCl salt. It will be noted that the value
of the AUC at
ED50 in Run # 2 for the hydrochloride salt of Compound A (shown in Table 5) is
in obvious
disagreement with the data obtained from the two other runs performed with the
hydrochloride
salt of Compound A as well as the run performed with the free base. The reason
for this
discrepancy is unknown, and we do not believe that the pharmacokinetic data
obtained in Run
# 2 (Table 5) are representative of the characteristics of Compound A.

14


CA 02703257 2010-04-20
WO 2009/058267 PCT/US2008/012224

A comparison of the average AUC at the ED50 for the 2 other studies conducted
with
the Compound A HC1 salt (i.e., Tables 4 and 6) and the study conducted with
the Compound
A free base (Table 3) is similar to the value obtained from Run #3 of the HCl
salt reported in
Table 6 (313 ng*hr/ml vs. 338 ng*hr/ml). Consequently the data from Run #3 are
used to
represent the efficacy of compound A in the rat corneal angiogenesis model:
ED50 = 0.14
mg/kg, AUC at ED50=338 ng*hr/ml.

Table 7 provides additional information about the pharmacokinetic profiles of
reference
compound I and Compound A. The data contained in this table was obtained using
methods
well known in the art.

TABLE 7
PK Parameter Ref. Cmpd 1 Cmpd A
Rat Clearance (L/h/Kg) 0.8 0.4
Rat Volume of Distribution (L/Kg) 1.0 1.0

Rat I.V. T1/2 (h) 4.73 2.1
Rat P.O. Ti,2 (h) 2.4 2.0
Rat P.O. (2mpk) %F 67 59
Rat P.O. (2mpk) AUCo_, (ng*h/ml) 1705 2796
Rat P.O. (2mpk) AUCo_;ff (ng*h/ml) 1706 2813
Rat Liver Microsome ( L/min/mg) 65 45
Human Liver Microsome ( L/min/mg) 29 17

Tables 8 - 14 below provide results obtained in various in vivo xenograft
tumor models
using Compound A. The procedures used are well known in the art. In general,
tumor cell
lines of interest are expanded in culture, harvested and injected
subcutaneously into 5-8 week
old female nude mice (CD 1 nu/nu, Charles River Labs) (n = 10). Subsequent
administration of
compound by oral gavage begins anywhere from day 10 to day 28 post tumor cell
challenge
and continues once a day for the duration of the experiment. Progression of
tumor growth is
followed by three dimensional caliper measurements and recorded as a function
of time. Initial
statistical analysis is done by repeated measures analysis of variance
(RMANOVA) followed
by Scheffe post hoc testing for multiple comparisons. Vehicle alone (Ora-
PlusTM) was used as
the control.



CA 02703257 2010-04-20
WO 2009/058267 PCT/US2008/012224
TABLE 8
Effect of Compound A (po, qd) on HT29 Xenografts
Treatment Tumor Volume mm / Standard Error
Group day 14 day 17 day 20 day 25 day 29
Vehicle 250/ 17 367 / 34 545 / 51 715 / 69 869 / 91
3 mpk Cmpd A 248 / 16 343 /38 450 / 38 502 / 48 499 / 52
m k Cm pd A 249 / 16 313 /22 374 /35 396 / 39 326 / 30
30 m k Cm pd A 250/ 17 290/ 13 354 / 26 286 / 23 248 / 19
5
TABLE 9
Effect of Compound A (po, qd) on A431 Xenografts
Treatment Tumor Volume mm / Standard Error
Group day 11 day 14 day 17 day 21 day 24
Vehicle 266 / 18 366 / 48 518 / 76 734 / 100 1036 / 79
1 m k Cm pd A 264 / 16 396 / 21 547 / 37 850 / 75 622 / 47
3 mpk Cm pd A 267 / 19 366 / 37 461 / 30 550 / 44 390 / 20
10m kCmpdA 267/18 311/30 298/33 248/30 179/23.
TABLE 10

Effect of Compound A (po, qd) on Large Established Calu-6 Xenografts
Treatment Tumor Volume mm / Standard Error
Group day 28 day 32 day 35 day 39 day 42 day 45 day 48
10 mpk 705 / 54 761 / 58 731 / 53 791 / 55 834 / 65 963 / 78 967 / 91
Cm pd A

TABLE 11

Effect of Compound A (po, qd) on Large Established A431 Xenografts
Treatment Tumor Volume mm / Standard Error
Group day day day day day day day day day
16 20 23 27 30 33 36 47 50
10 mpk 633/ 539/ 388/ 335/ 274/ 236/ 240/ 226/ 217/
Cm pd A 41 41 38 33 39 42 45 39 42
16


CA 02703257 2010-04-20
WO 2009/058267 PCT/US2008/012224
TABLE 12
Effect of Compound A (po, qd) on Calu-6 Xenografts
Treatment Tumor Volume mm / Standard Error
Group day 17 day 20 day 24 day 28 day 31
Vehicle 283 / 22 463 / 33 588 / 44 774 / 73 954 / 84
1 m k Cm pd A 282 / 23 428 / 36 528 / 45 599 / 52 697 / 64
3 m k Cm pd A 283 / 24 390 / 38 455 / 42 507 53 588 / 66
mkCm Cm285/24 369/37 401 /56 442/64 534/68

5
TABLE 13
Effect of Compound A (po, qd) on A431 (MOA) Xenografts
Treatment Tumor Volume mm 3)/s andard Error
Group t= 0 t=12hr day 1 day 2 day 3 day 7 day 14
Vehicle 223 / 24 241 / 25 269 / 29 335 / 42 365 / 41 528 / 49 760 / 80
10 mpk 228 / 22 227/ 18 243 / 28 225 / 24 232 / 28 183 / 30
Cm pd A

TABLE 14
Effect of Compound A (po, qd) on A431 (MOA) Xenografts
Treatment Tumor Volume mm) / Standard Error
Group t=0 t=6hr t=12hr day day day day day day
1 2 3 5 7 14
Vehicle 351 / 359/ 370/ 385/ 404/ 435/ 599/ 760/ 597/
26 36 37 45 26 36 45 80 44
30 mpk 342/ 320/ 348/ 333/ 314/ 300/ 183 / 268/
Cmpd A 23 21 31 33 28 19 30 21
Table 15 below provides the results obtained when Compound A was run in an in
vivo
bone metastasis model. The procedure used to generate the data is as follows:

Materials and Methods
MDA-231 Luc cells (1 x 105) were injected into the cardiac left ventricle of 4-
to 6-week old,
female, athymic nude mice (Harlan Sprague Dawley). Positive intra-cardiac
injections were
confirmed by whole body bioluminescence, and mice were randomized into groups
(n = 10).
Treatment began on day 0 with either of the following 4 treatments: Ora-Plus
PO as a vehicle
twice daily, recombinant OPG (OPG-Fc) 3.0 mg/kg SC three times weekly,
Compound A 30
mg/kg PO twice daily, and Compound A PO once daily. In vivo bioluminescent
imaging was
performed twice weekly with an IVIS-200 imaging system (Xenogen Corp.).
Fifteen minutes
prior to imaging, mice were given 150 mg/kg luciferin by i.p. injection.
Images were collected
17


CA 02703257 2010-04-20
WO 2009/058267 PCT/US2008/012224
and analyzed with Living Image software (Xenogen Corp.), with the region of
interest
including the femur/tibia region of the hind limbs.

TABLE 15
Treatment Bioluminescence (photons/sec) / Standard Error
Group Day 1 Day 5 Day 8 Day 13 Day 15 Day 19 Day 22
Vehicle 49.94 / 5.32 / 5.93 / 6.80 / 7.38 / 7.94 / 8.60 /
0.11 0.14 0.18 0.17 0.18 0.18 0.24

OPG-Fc 3 mg/kg 4.80 / 5.40 / 5.61 / 6.75 / 7.23 / 8.00 / 8.42 /
0.07 0.11 0.15 0.14 0.14 0.12 0.10
Cmpd A 5.20 / 5.24 / 6.21 / 6.87 / 7.36 / 7.88 / 7.95 /
30 mg/kg BID 0.09 0.12 0.10 0.14 0.16 0.14 0.22
Cmpd A 5.06/ 5.43/ 6.12/ 7.03/ 7.55/ 7.99/ 8.50/
30 mg/kg QD 0.08 0.11 0.08 0.16 0.12 0.20 0.15
INDICATIONS
Compounds of the present invention would be useful for, but not limited to,
the
prevention or treatment of angiogenesis related diseases. The compounds of the
invention
have kinase inhibitory activity, such as VEGFR/KDR inhibitory activity. The
compounds of
the invention are useful in therapy as antineoplasia agents or to minimize
deleterious effects of
VEGF.
Compounds of the invention would be useful for the treatment of neoplasia
including
cancer and metastasis, including, but not limited to: carcinoma such as cancer
of the bladder,
breast, colon, kidney, liver, lung (including small cell lung cancer),
esophagus, gall-bladder,
ovary, pancreas, stomach, cervix, thyroid, prostate, and skin (including
squamous cell
carcinoma); hematopoietic tumors of lymphoid lineage (including leukemia,
acute lymphocitic
leukemia, acute lymphoblastic leukemia, B-cell lymphoma, T-cell-lymphoma,
Hodgkin's
lymphoma, non-Hodgkin's lymphoma, hairy cell lymphoma and Burkett's lymphoma);
hematopoietic tumors of myeloid lineage (including acute and chronic
myelogenous
leukemias, myelodysplastic syndrome and promyelocytic leukemia); tumors of
mesenchymal
origin (including fibrosarcoma and rhabdomyosarcoma, and other sarcomas, e.g.
soft tissue
and bone); tumors of the central and peripheral nervous system (including
astrocytoma,
neuroblastoma, glioma and schwannomas); and other tumors (including melanoma,
seminoma,
18


CA 02703257 2010-04-20
WO 2009/058267 PCT/US2008/012224
teratocarcinoma, osteosarcoma, xenoderoma pigmentosum, keratoctanthoma,
thyroid follicular
cancer and Kaposi's sarcoma).
Preferably, the compounds are useful for the treatment of neoplasia selected
from lung
cancer, colon cancer and breast cancer.
The compounds also would be useful for treatment of ophthalmological
conditions such
as corneal graft rejection, ocular neovascularization, retinal
neovascularization including
neovascularization following injury or infection, diabetic retinopathy,
retrolental fibroplasia
and neovascular glaucoma; retinal ischemia; vitreous hemorrhage; ulcerative
diseases such as
gastric ulcer; pathological, but non-malignant, conditions such as
hemangiomas, including
infantile hemaginomas, angiofibroma of the nasopharynx and avascular necrosis
of bone; and
disorders of the female reproductive system such as endometriosis. The
compounds are also
useful for the treatment of edema, and conditions of vascular
hyperpermeability.
The compounds of the invention are useful in therapy of proliferative
diseases. These
compounds can be used for the treatment of an inflammatory rheumatoid or
rheumatic disease,
especially of manifestations at the locomotor apparatus, such as various
inflammatory
rheumatoid diseases, especially chronic polyarthritis including rheumatoid
arthritis, juvenile
arthritis or psoriasis arthropathy; paraneoplastic syndrome or tumor-induced
inflammatory
diseases, turbid effusions, collagenosis, such as systemic Lupus
erythematosus, poly-myositis,
dermato-myositis, systemic sclerodermia or mixed collagenosis; postinfectious
arthritis (where
no living pathogenic organism can be found at or in the affected part of the
body), seronegative
spondylarthritis, such as spondylitis ankylosans; vasculitis, sarcoidosis, or
arthrosis; or further
any combinations thereof. An example of an inflammation related disorder is
(a) synovial
inflammation, for example, synovitis, including any of the particular forms of
synovitis, in
particular bursal synovitis and purulent synovitis, as far as it is not
crystal-induced. Such
synovial inflammation may for example, be consequential to or associated with
disease, e.g.
arthritis, e.g. osteoarthritis, rheumatoid arthritis or arthritis deformans.
The present invention
is further applicable to the systemic treatment of inflammation, e.g.
inflammatory diseases or
conditions, of the joints or locomotor apparatus in the region of the tendon
insertions and
tendon sheaths. Such inflammation may be, for example, consequential to or
associated with
disease or further (in a broader sense of the invention) with surgical
intervention, including, in
particular conditions such as insertion endopathy, myofasciale syndrome and
tendomyosis.
The present invention is further especially applicable to the treatment of
inflammation, e.g.
inflammatory disease or condition, of connective tissues including
dermatomyositis and
myositis.

19


CA 02703257 2010-04-20
WO 2009/058267 PCT/US2008/012224
These compounds can be used as active agents against such disease states as
arthritis,
atherosclerosis, psoriasis, hemangiomas, myocardial angiogenesis, coronary and
cerebral
collaterals, ischemic limb angiogenesis, wound healing, peptic ulcer
Helicobacter related
diseases, fractures, cat scratch fever, rubeosis, neovascular glaucoma and
retinopathies such as
those associated with diabetic retinopathy or macular degeneration. In
addition, some of these
compounds can be used as active agents against solid tumors, malignant
ascites, hematopoietic
cancers and hyperproliferative disorders such as thyroid hyperplasia
(especially Grave's
disease), and cysts (such as hypervascularity of ovarian stroma,
characteristic of polycystic
ovarian syndrome (Stein- Leventhal syndrome)) since such diseases require a
proliferation of
blood vessel cells for growth and/or metastasis.
Further, some of these compounds can be used as active agents against burns,
chronic
lung disease, stroke, polyps, anaphylaxis, chronic and allergic inflammation,
ovarian
hyperstimulation syndrome, brain tumor-associated cerebral edema, high-
altitude, trauma or
hypoxia induced cerebral or pulmonary edema, ocular and macular edema,
ascites, and other
diseases where vascular hyperpermeability, effusions, exudates, protein
extravasation, or
edema is a manifestation of the disease. The compounds will also be useful in
treating
disorders in which protein extravasation leads to the deposition of fibrin and
extracellular
matrix, promoting stromal proliferation (e.g. fibrosis, cirrhosis and carpal
tunnel syndrome).
The compounds of the present invention are also useful in the treatment of
ulcers
including bacterial, fungal, Mooren ulcers and ulcerative colitis.
The compounds of the present invention are also useful in the treatment of
conditions
wherein undesired angiogenesis, edema, or stromal deposition occurs in viral
infections such as
Herpes simplex, Herpes Zoster, AIDS, Kaposi's sarcoma, protozoan infections
and
toxoplasmosis, following trauma, radiation, stroke, endometriosis, ovarian
hyperstimulation
syndrome, systemic lupus, sarcoidosis, synovitis, Crohn's disease, sickle cell
anemia, Lyme
disease, pemphigoid, Paget's disease, hyperviscosity syndrome, Osler-Weber-
Rendu disease,
chronic inflammation, chronic occlusive pulmonary disease, asthma, and
inflammatory
rheumatoid or rheumatic disease. The compounds are also useful in the
reduction of sub-
cutaneous fat and for the treatment of obesity.
The compounds of the present invention are also useful in the treatment of
ocular
conditions such as ocular and macular edema, ocular neovascular disease,
scleritis, radial
keratotomy, uveitis, vitritis, myopia, optic pits, chronic retinal detachment,
post-laser
complications, glaucoma, conjunctivitis, Stargardt's disease and Eales disease
in addition to
retinopathy and macular degeneration.



CA 02703257 2010-04-20
WO 2009/058267 PCT/US2008/012224
The compounds of the present invention are also useful in the treatment of
cancer
related indications such as solid tumors, sarcomas (especially Ewing's sarcoma
and
osteosarcoma), retinoblastoma, rhabdomyosarcomas, neuroblastoma, hematopoietic
malignancies, including leukemia and lymphoma, tumor- induced pleural or
pericardial
effusions, and malignant ascites.
As used herein, the compounds of the present invention include the
pharmaceutically
acceptable derivatives thereof.
Where the plural form is used for compounds, salts, and the like, this is
taken to mean
also a sindle compound, salt and the like.

DEFINITIONS
"Angiogenesis" is defined as any alteration of an existing vascular bed or the
formation
of new vasculature, which benefits tissue perfasion. This includes the
formation of new
vessels by sprouting of endothelial cells from existing blood vessels or the
remodeling of
existing vessels to alter size, maturity, direction or flow properties to
improve blood perfusion
of tissue.
The terms "cancer" and "cancerous" when used herein refer to or describe the
physiological condition in mammals that is typically characterized by
unregulated cell growth.
Examples of cancer include but are not limited to, carcinoma, lymphoma,
sarcoma, blastoma
and leukemia. More particular examples of such cancers include squamous cell
carcinoma,
lung cancer, pancreatic cancer, cervical cancer, bladder cancer, hepatoma,
breast cancer, colon
carcinoma, and head and neck cancer. The terms "treating," "treatment," and
"therapy" as used
herein refer to curative therapy, prophylactic therapy, and preventative
therapy.
The term "mammal" as used herein refers to any mammal classified as a mammal,
including humans, cows, horses, dogs and cats. In a preferred embodiment of
the invention, the
mammal is a human.
The term "treatment" (or "treating") includes therapeutic treatment as well as
prophylactic treatment (either preventing the onset of disorders altogether or
delaying the onset
of a pre-clinically evident stage of disorders in individuals).
The phrase "therapeutically-effective" is intended to qualify the amount of
each agent,
which will achieve the goal of improvement in disorder severity and the
frequency of incidence
over treatment of each agent by itself, while avoiding adverse side effects
typically associated
with alternative therapies. For example, effective neoplastic therapeutic
agents prolong the

21


CA 02703257 2010-04-20
WO 2009/058267 PCT/US2008/012224
survivability of the patient, inhibit the rapidly proliferating cell growth
associated with the
neoplasm, or effect a regression of the neoplasm.
The term "halogen" (or "halo") means fluorine, chlorine, bromine or iodine
atoms.
The term "aryl", alone or in combination, means a carbocyclic aromatic system
containing one or two rings wherein such rings may be attached together in a
fused manner.
The term "aryl" embraces aromatic radicals such as phenyl, naphthyl, indenyl,
tetrahydronaphthyl, and indanyl. More preferred aryl is phenyl. Said "aryl"
group may have I
or more substituents such as lower alkyl, hydroxyl, halo, haloalkyl, nitro,
cyano, alkoxy, lower
alkylamino, and the like. Phenyl substituted with -O-CH2-O- forms the aryl
benzodioxolyl

substituent.
The term "heteroaryl" denotes aryl ring systems that contain one or more
heteroatoms
selected from the group 0, N and S, wherein the ring nitrogen and sulfur
atom(s) are optionally
oxidized, and nitrogen atom(s) are optionally quarternized. Examples include
unsaturated 5 to
6 membered heteromonocyclyl group containing I to 4 nitrogen atoms, for
example, pyrrolyl,
imidazolyl, pyrazolyl, 2-pyridyl, 3-pyridyl, 4-pyridyl, pyrimidyl, pyrazinyl,
pyridazinyl,
triazolyl [e.g., 4H-1,2,4-triazolyl, 1H-1,2,3-triazolyl, 2H-1,2,3-triazolyl];
unsaturated 5- to 6-
membered heteromonocyclic group containing an oxygen atom, for example,
pyranyl, 2-furyl,
3-furyl, etc.; unsaturated 5 to 6-membered heteromonocyclic group containing a
sulfur atom,
for example, 2-thienyl, 3-thienyl, etc.; unsaturated 5- to 6-membered
heteromonocyclic group
containing I to 2 oxygen atoms and I to 3 nitrogen atoms, for example,
oxazolyl, isoxazolyl,
oxadiazolyl [e.g., 1,2,4-oxadiazolyl, 1,3,4-oxadiazolyl, 1,2,5-oxadiazolyl];
unsaturated 5 to 6-
membered heteromonocyclic group containing 1 to 2 sulfur atoms and 1 to 3
nitrogen atoms,
for example, thiazolyl, thiadiazolyl [e.g., 1,2,4-thiadiazolyl, 1,3,4-
thiadiazolyl, 1,2,5-
thiadiazolyl]. Said "heteroaryl" group may have I or more substituents such as
lower alkyl,
hydroxyl, halo, haloalkyl, nitro, cyano, alkoxy, lower alkylamino, and the
like.
The term "comprising" is meant to be open ended, including the indicated
component
but not excluding other elements.

22


CA 02703257 2010-04-20
WO 2009/058267 PCT/US2008/012224
The term "active metabolite" as used herein refers any of the following
compounds:
H
Oy N N

O O N
/ N
Y N\O
N

O
O / N/
O HO N

H H
N N
N\O OYN /N
\O
N

\ - OH
\
HO

N/ and N
The present invention also comprises the use of a compound of the invention,
or
pharmaceutically acceptable salt thereof, in the manufacture of a medicament
for the treatment
either acutely or chronically of an angiogenesis mediated disease state,
including those
described previously. The compounds of the present invention are useful in the
manufacture of
an anti-cancer medicament.
The present invention comprises a pharmaceutical composition comprising a
therapeutically effective amount of a compound of the current invention in
association with a
least one pharmaceutically acceptable carrier, adjuvant or diluent.
The present invention also comprises a method of treating angiogenesis related
disorders in a subject having or susceptible to such disorder, the method
comprising treating
the subject with a therapeutically effective amount of a compound of the
current invention.
COMBINATIONS
While the compounds of the invention can be administered as the sole active
pharmaceutical agent, they can also be used in combination with one or more
compounds of
the invention or other agents. When administered as a combination, the
therapeutic agents can
be formulated as separate compositions that are administered at the same time
or sequentially
at different times, or the therapeutic agents can be given as a single
composition.
The phrase "co-therapy" (or "combination-therapy"), in defining use of a
compound of
the present invention and another pharmaceutical agent, is intended to embrace
administration
23


CA 02703257 2010-04-20
WO 2009/058267 PCT/US2008/012224
of each agent in a sequential manner in a regimen that will provide beneficial
effects of the
drug combination, and is intended as well to embrace co-administration of
these agents in a
substantially simultaneous manner, such as in a single capsule having a fixed
ratio of these
active agents or in multiple, separate capsules for each agent.
Specifically, the administration of compounds of the present invention may be
in
conjunction with additional therapies known to those skilled in the art in the
prevention or
treatment of neoplasia, such as with radiation therapy or with cytostatic or
cytotoxic agents.
If formulated as a fixed dose, such combination products employ the compounds
of this
invention within the accepted dosage ranges. Compounds of the current
invention may also be
administered sequentially with known anticancer or cytotoxic agents when a
combination
formulation is inappropriate. The invention is not limited in the sequence of
administration;
compounds of the invention may be administered either prior to, simultaneous
with or after
administration of the known anticancer or cytotoxic agent.
Currently, standard treatment of primary tumors consists of surgical excision
followed
by either radiation or IV administered chemotherapy. The typical chemotherapy
regime
consists of either DNA alkylating agents, DNA intercalating agents, CDK
inhibitors, or
microtubule poisons. The chemotherapy doses used are just below the maximal
tolerated dose
and therefore dose limiting toxicities typically include, nausea, vomiting,
diarrhea, hair loss,
neutropenia and the like.
There are large numbers of antineoplastic agents available in commercial use,
in
clinical evaluation and in pre-clinical development, which would be selected
for treatment of
neoplasia by combination drug chemotherapy. Such antineoplastic agents fall
into several
major categories, namely, antibiotic-type agents, alkylating agents,
antimetabolite agents,
hormonal agents, immunological agents, interferon-type agents and a category
of
miscellaneous agents.
A first family of antineoplastic agents, which may be used in combination with
compounds of the present invention, consists of antimetabolite-
type/thymidiIate synthase
inhibitor antineoplastic agents. Suitable antimetabolite antineoplastic agents
may be selected
from but not limited to the group consisting of 5-FU-fibrinogen, acanthifolic
acid,
aminothiadiazole, brequinar sodium, carmofur, Ciba-Geigy CGP-30694,
cyclopentyl cytosine,
cytarabine phosphate stearate, cytarabine conjugates, Lilly DATHF, Merrel Dow
DDFC,
dezaguanine, dideoxycytidine, dideoxyguanosine, didox, Yoshitomi DMDC,
doxifluridine,
Wellcome EHNA, Merck & Co. EX-015, fazarabine, floxuridine, fludarabine
phosphate, 5-
fluorouracil, N-(2'-furanidyl)-5-fluorouracil, Daiichi Seiyaku FO- 152,
isopropyl pyrrolizine,

24


CA 02703257 2010-04-20
WO 2009/058267 PCT/US2008/012224
Lilly LY-188011, Lilly LY-264618, methobenzaprim, methotrexate, Wellcome
MZPES,
norspennidine, NCI NSC-127716, NCI NSC-264880, NCI NSC-39661, NCI NSC-612567,
Warner-Lambert PALA, pentostatin, piritrexim, plicamycin, Asahi Chemical PL-
AC, Takeda
TAC-788, thioguanine, tiazofurin, Erbamont TIF, trimetrexate, tyrosine kinase
inhibitors,
Taiho UFT and uricytin.
A second family of antineoplastic agents, which may be used in combination
with
compounds of the present invention, consists of alkylating-type antineoplastic
agents. Suitable
alkylating-type antineoplastic agents may be selected from but not limited to
the group
consisting of Shionogi 254-S, aldo-phosphamide analogues, altretamine,
anaxirone, Boehringer
Mannheim BBR-2207, bestrabucil, budotitane, Wakunaga CA-102, carboplatin,
carmustine,
Chinoin- 139, Chinoin- 153, chlorambucil, cisplatin, cyclophosphamide,
American Cyanamid
CL-286558, Sanofi CY-233, cyplatate, Degussa D-19-384, Sumimoto DACHP(Myr)2,
diphenylspiromustine, diplatinum cytostatic, Erba distamycin derivatives,
Chugai DWA-
2114R, ITI E09, elmustine, Erbamont FCE-24517, estramustine phosphate sodium,
fotemustine, Unimed G-6-M, Chinoin GYKI-17230, hepsul-fam, ifosfamide,
iproplatin,
lomustine, mafosfamide, mitolactol, Nippon Kayaku NK-121, NCI NSC-264395, NCI
NSC-
342215, oxaliplatin, Upjohn PCNU, prednimustine, Proter PTT-119, ranimustine,
semustine,
SmithKline SK&F-101772, Yakult Honsha SN-22, spiromus-tine, Tanabe Seiyaku TA-
077,
tauromustine, temozolomide, teroxirone, tetraplatin and trimelamol.
A third family of antineoplastic agents which may be used in combination with
compounds of the present invention consists of antibiotic-type antineoplastic
agents. Suitable
antibiotic-type antineoplastic agents may be selected from but not limited to
the group
consisting of Taiho 4181-A, aclarubicin, actinomycin D, actinoplanone,
Erbamont ADR-456,
aeroplysinin derivative, Ajinomoto AN-201-II, Ajinomoto AN-3, Nippon Soda
anisomycins,
anthracycline, azino-mycin-A, bisucaberin, Bristol-Myers BL-6859, Bristol-
Myers BMY-
25067, Bristol-Myers BMY-25551, Bristol-Myers BMY-26605, Bristol-Myers BMY-
27557,
Bristol-Myers BMY-28438, bleomycin sulfate, bryostatin-1, Taiho C-1027,
calichemycin,
chromoximycin, dactinomycin, daunorubicin, Kyowa Hakko DC-102, Kyowa Hakko DC-
79,
Kyowa Hakko DC-88A, Kyowa Hakko DC89-A1, Kyowa Hakko DC92-B, ditrisarubicin B,
Shionogi DOB-41, doxorubicin, doxorubicin-fibrinogen, elsamicin-A, epirubicin,
erbstatin,
esorubicin, esperamicin-A1, esperamicin-Alb, Erbamont FCE-21954, Fujisawa FK-
973,
fostriecin, Fujisawa FR-900482, glidobactin, gregatin-A, grincamycin,
herbimycin, idarubicin,
illudins, kazusamycin, kesarirhodins, Kyowa Hakko KM-5539, Kirin Brewery KRN-
8602,
Kyowa Hakko KT-5432, Kyowa Hakko KT-5594, Kyowa Hakko KT-6149, American



CA 02703257 2010-04-20
WO 2009/058267 PCT/US2008/012224
Cyanamid LL-D49194, Meiji Seika ME 2303, menogaril, mitomycin, mitoxantrone,
SmithKline M-TAG, neoenactin, Nippon Kayaku NK-313, Nippon Kayaku NKT-01, SRI
International NSC-357704, oxalysine, oxaunomycin, peplomycin, pilatin,
pirarubicin,
porothramycin, pyrindanycin A, Tobishi RA-I, rapamycin, rhizoxin, rodorubicin,
sibanomicin,
siwenmycin, Sumitomo SM-5887, Snow Brand SN-706, Snow Brand SN-07, sorangicin-
A,
sparsomycin, SS Pharmaceutical SS-21020, SS Pharmaceutical SS-7313B, SS
Pharmaceutical
SS-9816B, steffimycin B, Taiho 4181-2, talisomycin, Takeda TAN-868A,
terpentecin,
thrazine, tricrozarin A, Upjohn U-73975, Kyowa Hakko UCN-10028A, Fujisawa WF-
3405,
Yoshitomi Y-25024 and zorubicin.
A fourth family of antineoplastic agents which may be used in combination with
compounds of the present invention consists of a miscellaneous family of
antineoplastic
agents, including tubulin interacting agents, topoisomerase II inhibitors,
topoisomerase I
inhibitors and hormonal agents, selected from but not limited to the group
consisting of a-
carotene, a-difluoromethyl-arginine, acitretin, Biotec AD-5, Kyorin AHC-52,
alstonine,

amonafide, amphethinile, amsacrine, Angiostat, ankinomycin, anti-neoplaston
A10,
antineoplaston A2, antineoplaston A3, antineoplaston AS, antineoplaston AS2-1,
Henkel APD,
aphidicolin glycinate, asparaginase, Avarol, baccharin, batracylin, benfluron,
benzotript, Ipsen-
Beaufour BIM-23015, bisantrene, Bristol-Myers BMY-40481, Vestar boron-10,
bromofosfainide, Wellcome BW-502, Wellcome BW-773, caracemide, carmethizole
hydrochloride, Ajinomoto CDAF, chlorsulfaquinoxalone, Cherries CHX-2053,
Chemex CHX-
100, Warner-Lambert CI-921, Warner-Lambert CI-937, Warner-Lambert CI-941,
Warner-
Lambert CI-958, clanfenur, claviridenone, ICN compound 1259, ICN compound
4711,
Contracan, Yakult Honsha CPT-11, crisnatol, curaderm, cytochalasin B,
cytarabine, cytocytin,
Merz D-609, DABIS maleate, dacarbazine, datelliptinium, didemnin-B,
dihaematoporphyrin
ether, dihydrolenperone, dinaline, distamycin, Toyo Pharmar DM-341, Toyo
Pharmar DM-75,
Daiichi Seiyaku DN-9693, docetaxel elliprabin, elliptinium acetate, Tsumura
EPMTC, the
epothilones, ergotamine, etoposide, etretinate, fenretinide, Fujisawa FR-
57704, gallium nitrate,
genkwadaphnin, Chugai GLA-43, Glaxo GR-63178, grifolan NMF-5N,
hexadecylphosphocholine, Green Cross HO-221, homoharringtonine, hydroxyurea,
BTG
ICRF-187, ilmofosine, isoglutamine, isotretinoin, Otsuka JI-36, Ramot K-477,
Otsuak K-
76000Na, Kureha Chemical K-AM, MECT Corp KI-81 10, American Cyanamid L-623,
leukoregulin, lonidamine, Lundbeck LU-23-112, Lilly LY-186641, NCI (US) MAP,
marycin,
Merrel Dow MDL-27048, Medco MEDR-340, merbarone, merocyanlne derivatives,
methylanilinoacridine, Molecular Genetics MGI- 136, minactivin, mitonafide,
mitoquidone

26


CA 02703257 2012-03-13

WO 2009/058267 PCT/US2008/012224
mopidamol, motretinide, Zenyaku Kogyo MST-16, N-(retinoyl)amino acids, Nisshin
Flour
Milling N-021, N-acylated-dehydroalanines, nafazatrom, Taisho NCU-190,
nocodazole
derivative, Normosang, NCI NSC-145813, NCI NSC-361456, NCI NSC-604782, NCI NSC-

95580, ocreotide, Ono ONO-112, oquizanocine, Akzo Org-10172, paclitaxel,
pancratistatin,
pazelliptine, Warner-Lambert PD-111707, Warner-Lambert PD-115934, Warner-
Lambert PD-
131141, Pierre Fabre PE-1001, ICRT peptide D, piroxantrone,
polyhaematoporphyrin,
polypreic acid, Efamol porphyrin, probimane, procarbazine, proglumide,
Invitron protease
nexin I, Tobishi RA-700, razoxane, Sapporo Breweries RBS, restrictin-P,
retelliptine, retinoic
acid, Rhone-Poulenc RP-49532, Rhone-Poulenc RP-56976, SmithKline SK&F-104864,
Sumitomo SM- 108, Kuraray SMANCS, SeaPharm SP- 10094, spatol,
spirocyclopropane
derivatives, spirogermanium, Unimed, SS Pharmaceutical SS-554, strypoldinone,
Stypoldione,
Suntory SUN 0237, Suntory SUN 2071, superoxide dismutase, Toyama T-506, Toyama
T-680,
taxol, Teijin TEI-0303, teniposide, thaliblastine, Eastman Kodak TJB-29,
tocotrienol,
topotecan, Topostin, Teijin TT-82, Kyowa Hakko UCN-01, Kyowa Hakko UCN-1028,
ukrain,
Eastman Kodak USB-006, vinblastine sulfate, vincristine, vindesine,
vinestramide, vinorelbine,
vintriptol, vinzolidine, withanolides and Yamanouchi YM-534.
Alternatively, the present compounds may also be used in co-therapies with
other anti-
neoplastic agents, such as acemannan, aclarubicin, aldesleukin, alemtuzumab,
alitretinoin,
altretamine, amifostine, aminolevulinic acid, amrubicin, amsacrine,
anagrelide, anastrozole,
ANCER, ancestim, ARGLABIN, arsenic trioxide, BAM 002 (Novelos), bexarotene,
bicalutamide, broxuridine, capecitabine, celmoleukin, cetrorelix, cladribine,
clotrimazole,
cytarabine ocfosfate, DA 3030 (Dong-A), daclizumab, denileukin diftitox,
deslorelin,
dexrazoxane, dilazep, docetaxel, docosanol, doxercalciferol, doxifluridine,
doxorubicin,
bromocriptine, carmustine, cytarabine, fluorouracil, HIT diclofenac,
interferon alfa,
daunorubicin, doxorubicin, tretinoin, edelfosine, edrecolomab, eflornithine,
emitefur,
epirubicin, epoetin beta, etoposide phosphate, exemestane, exisulind,
fadrozole, filgrastim,
finasteride, fludarabine phosphate, formestane, fotemustine, gallium nitrate,
gemcitabine,
gemtuzumab zogamicin, gimeracil/oteracil/tegafur combination, glycopine,
goserelin,
heptaplatin, human chorionic gonadotropin, human fetal alpha fetoprotein,
ibandronic acid,
idarubicin, (imiquirnod, interferon alfa, interferon alfa, natural, interferon
alfa-2, interferon
alfa-2a, interferon alfa-2b, interferon alfa-N 1, interferon alfa-n3,
interferon alfacon- 1,
interferon alpha, natural, interferon beta, interferon beta-I a, interferon
beta-I b, interferon
gamma, natural interferon gamma-I a, interferon gamma-I b, interleukin- I
beta, iobenguane,
irinotecan, irsogladine, lanreotide, LC 9018 (Yakult), leflunomide,
lenograstim, lentinan

27
*Trademark


CA 02703257 2010-04-20
WO 2009/058267 PCT/US2008/012224
sulfate, letrozole, leukocyte alpha interferon, leuprorelin, levamisole +
fluorouracil, liarozole,
lobaplatin, lonidamine, lovastatin, masoprocol, melarsoprol, metoclopramide,
mifepristone,
miltefosine, mirimostim, mismatched double stranded RNA, mitoguazone,
mitolactol,
mitoxantrone, molgramostim, nafarelin, naloxone + pentazocine, nartograstim,
nedaplatin,
nilutainide, noscapine, novel erythropoiesis stimulating protein, NSC 631570
octreotide,
oprelvekin, osaterone, oxaliplatin, paclitaxel, pamidronic acid, pegaspargase,
peginterferon
alfa-2b, pentosan polysulfate sodium, pentostatin, picibanil, pirarubicin,
rabbit antithymocyte
polyclonal antibody, polyethylene glycol interferon alfa-2a, porfimer sodium,
raloxifene,
raltitrexed, rasburicase, rhenium Re 186 etidronate, RII retinamide,
rituximab, romurtide,
samarium (153 Sm) lexidronam, sargramostim, sizofiran, sobuzoxane, sonermin,
strontium-89
chloride, surainin, tasonermin, tazarotene, tegafur, temoporfin, temozolomide,
teniposide,
tetrachlorodecaoxide, thalidomide, thymalfasin, thyrotropin alfa, topotecan,
toremifene,
tositumomab-iodine 131, trastuzumab, treosulfan, tretinoin, trilostane,
trimetrexate, triptorelin,
tumor necrosis factor alpha, natural, ubenimex, bladder cancer vaccine,
Maruyama vaccine,
melanoma lysate vaccine, valrubicin, verteporfin, vinorelbine, VIRULIZIN,
zinostatin
stimalamer, or zoledronic acid; abarelix; AE 941 (Aeterna), ambamustine,
antisense
oligonucleotide, bcl-2 (Genta), APC 8015 (Dendreon), cetuximab, decitabine,
dexaminoglutethimide, diaziquone, EL 532 (Elan), EM 800 (Endorecherche),
eniluracil,
etanidazole, fenretinide, filgrastim SDOI (Amgen), fulvestrant, galocitabine,
gastrin 17
immunogen, HLA-B7 gene therapy (Vical), granulocyte macrophage colony
stimulating
factor, histamine dihydrochloride, ibritumomab tiuxetan, ilomastat, IM 862
(Cytran),
interleukin-2, iproxifene, LDI 200 (Milkhaus), leridistim, lintuzumab, CA 125
MAb (Biomira),
cancer MAb (Japan Pharmaceutical Development), HER-2 and Fc MAb (Medarex),
idiotypic
105AD7 MAb (CRC Technology), idiotypic CEA MAb (Trilex), LYM-1-iodine 131 MAb
(Techniclone), polymorphic epithelial mucin-yttrium 90 MAb (Antisoma),
marimastat,
menogaril, mitumomab, motexafin gadolinium, MX 6 (Galderma), nelarabine,
nolatrexed, P 30
protein, pegvisomant, pemetrexed, porfiromycin, prinomastat, RL 0903 (Shire),
rubitecan,
satraplatin, sodium phenylacetate, sparfosic acid, SRL 172 (SR Pharma), SU
5416 (SUGEN),
TA 077 (Tanabe), tetrathiomolybdate, thaliblastine, thrombopoietin, tin ethyl
etiopurpurin,
tirapazamine, cancer vaccine (Biomira), melanoma vaccine (New York
University), melanoma
vaccine (Sloan Kettering Institute), melanoma oncolysate vaccine (New York
Medical
College), viral melanoma cell lysates vaccine (Royal Newcastle Hospital), or
valspodar.
Alternatively, the present compounds may also be used in-co-therapies with
VEGFR
inhibitors including

28


CA 02703257 2010-04-20
WO 2009/058267 PCT/US2008/012224
N-(4-chlorophenyl)-4-(4-pyridinylmethyl)-1-phthalazinamine;
4- [4- [ [ [ [4-chl oro -3 -(tri fl uoromethyl)phenyl]amino] carbonyl] amino]
phenoxy]-N-methyl -2-
pyridinecarboxamide;
N-[2-(diethyl amino)ethyl]-5-[(5-fluoro-l,2-dihydro-2-oxo-3H-indol-3-
ylidene)methyl]-2,4-
dimethyl-1 H-pyrrole-3 -carboxamide;
3 -[(4-bromo-2,6-difluorophenyl)methoxy] -5-[[ [ [4-(1-
pyrrolidinyl)butyl] amino] carbonyl] amino] -4-isothiazolecarboxamide;
N-(4-bromo-2-fluorophenyl)-6-methoxy-7-[(1-methyl-4-piperidinyl)methoxy]-4-
quinazolinamine;
3-[5,6,7,13-tetrahydro-9-[(1-methylethoxy)methyl]-5-oxo-12H-indeno[2,1-
a]pyrrolo[3,4-
c]carbazol- l 2-yl]propyl ester N,N-dimethyl-glycine;
N-[5- [ [[5-( 1,1 -dimethylethyl)-2-oxazolyl]methyl]thio]-2-thiazolyl]-4-
piperidinecarboxamide;
N-[3-chloro-4-[(3-fluorophenyl)methoxy]phenyl]-6-[5-[[[2-
(methylsulfonyl)ethyl]amino]methyl]-2-furanyl]-4-quinazolinamine

4-[(4-Methyl-l-piperazinyl)methyl]-N-[4-methyl-3-[[4-(3-pyridinyl)-2-
pyrimidinyl]amino] -
ph enyl]benzamide

N-(3-chloro-4-fluorophenyl)-7-methoxy-6-[3-(4-morpholinyl)propoxy]-4-
quinazolinamine
N-(3-ethynylphenyl)-6,7-bis(2-methoxyethoxy)-4-quinazolinamine
N-(3-((((2R)-1-methyl-2-pyrrolidinyl)methyl)oxy)-5-(trifluoromethyl)phenyl)-2-
((3-(1,3-
oxazol-5-yl)phenyl)amino)-3-pyridinecarboxamide;
2-(((4-fluorophenyl)methyl)amino)-N-(3-((((2R)-1-methyl-2-
pyrrolidinyl)methyl)oxy)-5-
(trifluoromethyl)phenyl)-3-pyridinecarboxamide;

N-[3 -(Azetidin-3-ylmethoxy)-5-trifluoromethyl-phenyl]-2-(4-fluoro-
benzylamino)-
nicotinamide.
6-fluoro-N-(4-(1-methylethyl)phenyl)-2-((4-pyridinylmethyl)amino)-3-
pyridinecarboxamide;
2-((4-pyridinylmethyl)amino)-N-(3-(((2S)-2-pyrrolidinylmethyl)oxy)-5-
(trifluoromethyl)phenyl)-3-pyridinecarboxamide;
N-(3-(1,1-dimethyl ethyl)-1 H-pyrazol-5-yl)-2-((4-pyridinylmethyl)aniino)-3-
pyridinecarboxamide;
N-(3,3-dimethyl-2,3-dihydro- I -benzofuran-6-yl)-2-((4-pyridinylmethyl)amino)-
3-
pyridinecarboxamide;
N-(3-((((2S)-1-methyl -2-pyrrolidinyl)methyl)oxy)-5-(trifluoromethyl)phenyl)-2-
((4-
pyridinylmethyl)amino)-3-pyridinecarboxamide;
29


CA 02703257 2010-04-20
WO 2009/058267 PCT/US2008/012224
2-((4-pyridinylmethyl)amino)-N-(3-((2-(1-pyrrolidinyl)ethyl)oxy)-4-
(trifluoromethyl)phenyl)-
3 -pyridinecarboxamide;
N-(3,3-dimethyl-2,3-dihydro- I H-indol-6-yl)-2-((4-pyridinylmethyl)amino)-3-
pyridinecarboxamide;
N-(4-(pentafluoroethyl)-3-(((2S)-2-pyrrolidinylmethyl)oxy)phenyl)-2-((4-
pyridinylmethyl)amino)-3-pyridinecarboxamide;
N-(3 -((3 -azetidinylmethyl)oxy)-5-(tri fluoromethyl)phenyl)-2-((4-
pyridinylmethyl)amino) -3-
pyri dinecarbox ami de;
N-(3-(4-piperidinyloxy)-5-(trifluoromethyl)phenyl)-2-((2-(3-
pyridinyl)ethyl)amino)-3-
pyridinecarboxamide;
N-(4,4-dimethyl-1,2,3,4-tetrahydro-isoquinolin-7-yl)-2-(1 H-indazol-6-ylamino)-
nicotinamide;
2-(1 H-indazol-6-ylamino)-N-[3-(1-methylpyrrolidin-2-ylmethoxy)-5-
trifluoromethyl-phenyl]-
nicotinamide;
N-[ 1-(2-dimethylamino-acetyl)-3,3-dimethyl-2,3-dihydro-1 H-indol-6-yl]-2-(1 H-
indazol-6-
ylamino)-nicotinamide;
2-(1 H-indazol-6-ylan:ino)-N-[3-(pyrrolidin-2-ylmethoxy)-5-trifluoromethyl-
phenyl]-
nicotinamide;
N-(1-acetyl -3,3-dim ethyl -2,3-dihydro-1 H-indol-6-yl)-2-(1 H-indazol-6-
ylamino)-nicotinamide;
N-(4,4-dimethyl-l-oxo-1,2,3,4-tetrahydro-isoquinolin-7-yl)-2-(1 H-indazol-6-
ylamino)-
nicotinamide;
N-[4-(tert-butyl)-3 -(3-piperidylpropyl)phenyl] [2-(1 H-indazol-6-ylamino)(3 -
pyridyl)]carboxamide;
N-[5-(tert-butyl)isoxazol-3-yl][2-(IH-indazol-6-ylamino)(3-
pyridyl)]carboxamide; and
N-[4-(tert-butyl)phenyl][2-(1 H-indazol-6-ylamino)(3-pyridyl)]carboxamide.
Other compounds described in the following patents and patent applications can
be
used in combination therapy: US 6,258,812, US 2003/0105091, WO 01/37820, US
6,235,764,
WO 01/32651, US 6,630,500, US 6,515,004, US 6,713,485, US 5,521,184, US
5,770,599, US
5,747,498, WO 02/68406, WO 02/66470, WO 02/55501, WO 04/05279, WO 04/07481, WO
04/07458, WO 04/09784, WO 02/59110, WO 99/45009, WO 00/59509, WO 99/61422, US
5,990,141, WO 00/12089 and WO 00/02871.
In some embodiments, the combination comprises a composition of the present
invention in combination with at least one anti-angiogenic agent. Agents are
inclusive of, but
not limited to, in vitro synthetically prepared chemical compositions,
antibodies, antigen
binding regions, radionuclides, and combinations and conjugates thereof. An
agent can be an



CA 02703257 2010-04-20
WO 2009/058267 PCT/US2008/012224
agonist, antagonist, allosteric modulator, toxin or, more generally, may act
to inhibit or
stimulate its target (e.g., receptor or enzyme activation or inhibition), and
thereby promote cell
death or arrest cell growth.
Exemplary anti-tumor agents include HERCEPTINTM (trastuzumab), which may be
used to treat breast cancer and other forms of cancer, and RITUXANTM
(rituximab),
ZEVALINTM (ibritumomab tiuxetan), and LYMPHOCIDETM (epratuzumab), which may be
used to treat non-Hodgkin's lymphoma and other forms of cancer, GLEEVACTM
which may be
used to treat chronic myeloid leukemia and gastrointestinal stromal tumors,
and BEXXARTM
(iodine 131 tositumomab) which may be used for treatment of non-Hodgkins's
lymphoma.
Exemplary anti-angiogenic agents include KDR (kinase domain receptor)
inhibitory
agents (e.g., antibodies and antigen binding regions that specifically bind to
the kinase domain
receptor), anti-VEGF agents (e.g., antibodies or antigen binding regions that
specifically bind
VEGF, or soluble VEGF receptors or a ligand binding region thereof) such as
AVASTINTM or
VEGF-TRAPTM, and anti-VEGF receptor agents (e.g., antibodies or antigen
binding regions
that specifically bind thereto), EGFR inhibitory agents (e.g., antibodies or
antigen binding
regions that specifically bind thereto) such as ERBITUXTM (IMC-C225), and
VECTIBIXrM
(panitumumab) IRESSATM (gefitinib), TARCEVATM (erlotinib), anti-Angl and anti-
Ang2
agents (e.g., antibodies or antigen binding regions specifically binding
thereto or to their
receptors, e.g., Tie2/Tek), and anti-Tie2 kinase inhibitory agents. The
pharmaceutical
compositions of the present invention can also include one or more agents
(e.g., antibodies,
antigen binding regions, or soluble receptors) that specifically bind and
inhibit the activity of
growth factors, such as antagonists of hepatocyte growth factor (HGF, also
known as Scatter
Factor), and antibodies or antigen binding regions that specifically bind its
receptor "c-Met" as
well as small molecules inhibitors of the c-Met kinase activity.
Other anti-angiogenic agents include Campath, IL-8, B-FGF, Tek antagonists
(Ceretti
et al., US Publication No. 2003/0162712; US Patent No. 6,413,932), anti-TWEAK
agents (e.g.,
specifically binding antibodies or antigen binding regions, or soluble TWEAK
receptor
antagonists; see, Wiley, US Patent No. 6,727,225), ADAM distintegrin domain to
antagonize
the binding of integrin to its ligands (Fanslow et al., US Publication No.
2002/0042368),
specifically binding anti-eph receptor and/or anti-ephrin antibodies or
antigen binding regions
(US Patent Nos. 5,981,245; 5,728,813; 5,969,110; 6,596,852; 6,232,447;
6,057,124 and patent
family members thereof), and anti-PDGF-BB antagonists (e.g., specifically
binding antibodies
or antigen binding regions) as well as antibodies or antigen binding regions
specifically

31


CA 02703257 2010-04-20
WO 2009/058267 PCT/US2008/012224
binding to PDGF-BB ligands, and PDGFR kinase inhibitory agents (e.g.,
antibodies or antigen
binding regions that specifically bind thereto).
Additional anti-angiogenic/anti-tumor agents include: SD-7784 (Pfizer, USA);
cilengitide.(Merck KGaA, Germany, EPO 770622); pegaptanib octasodium, (Gilead
Sciences,
USA); Alphastatin, (BioActa, UK); M-PGA, (Celgene, USA, US 5712291);
ilomastat, (Arriva,
USA, US 5892112); emaxanib, (Pfizer, USA, US 5792783); vatalanib, (Novartis,
Switzerland); 2-methoxyestradiol, (EntreMed, USA); TLC ELL-12, (Elan,
Ireland); anecortave
acetate, (Alcon, USA); alpha-D148 Mab, (Amgen, USA); CEP-7055,(Cephalon, USA);
anti-
Vn Mab, (Crucell, Netherlands) DAC:antiangiogenic, (ConjuChem, Canada);
Angiocidin,
(InKine Pharmaceutical, USA); KM-2550, (Kyowa Hakko, Japan); SU-0879, (Pfizer,
USA);
CGP-79787, (Novartis, Switzerland, EP 970070); ARGENT technology, (Ariad,
USA);
YIGSR-Stealth, (Johnson & Johnson, USA); fibrinogen-E fragment, (BioActa, UK);
angiogenesis inhibitor, (Trigen, UK); TBC-1635, (Encysive Pharmaceuticals,
USA); SC-236,
(Pfizer, USA); ABT-567, (Abbott, USA); Metastatin, (EntreMed, USA);
angiogenesis
inhibitor, (Tripep, Sweden); maspin, (Sosei, Japan); 2-methoxyestradiol,
(Oncology Sciences
Corporation, USA); ER-68203-00, (IVAX, USA); Benefin, (Lane Labs, USA); Tz-93,
(Tsumura, Japan); TAN-1120, (Takeda, Japan); FR-111142, (Fujisawa, Japan, JP
02233610);
platelet factor 4, (RepliGen, USA, EP 407122); vascular endothelial growth
factor antagonist,
(Borean, Denmark); cancer therapy, (University of South Carolina, USA);
bevacizumab
(pINN), (Genentech, USA); angiogenesis inhibitors, (SUGEN, USA); XL 784,
(Exelixis,
USA); XL 647, (Exelixis, USA); MAb, alpha5beta3 integrin, second generation,
(Applied
Molecular Evolution, USA and Medlmmune, USA); gene therapy, retinopathy,
(Oxford
BioMedica, UK); enzastaurin hydrochloride (USAN), (Lilly, USA); CEP 7055,
(Cephalon,
USA and Sanofi-Synthelabo, France); BC 1, (Genoa Institute of Cancer Research,
Italy);
angiogenesis inhibitor, (Alchemia, Australia); VEGF antagonist, (Regeneron,
USA); rBPI 21
and BPI-derived antiangiogenic, (XOMA, USA); PI 88, (Progen, Australia);
cilengitide
(pINN), (Merck KGaA, German; Munich Technical University, Germany, Scripps
Clinic and
Research Foundation, USA); cetuximab (INN), (Aventis, France); AVE 8062,
(Ajinomoto,
Japan); AS 1404, (Cancer Research Laboratory, New Zealand); SG 292, (Telios,
USA);
Endostatin, (Boston Childrens Hospital, USA); ATN 161, (Attenuon, USA);
ANGIOSTATIN,
(Boston Childrens Hospital, USA); 2-methoxyestradiol, (Boston Childrens
Hospital, USA); ZD
6474, (AstraZeneca, UK); ZD 6126, (Angiogene Pharmaceuticals, UK); PPI 2458,
(Praecis,
USA); AZD 9935, (AstraZeneca, UK); AZD 2171, (AstraZeneca, UK); vatalanib
(pINN),
(Novartis, Switzerland and Schering AG, Germany); tissue factor pathway
inhibitors,

32


CA 02703257 2010-04-20
WO 2009/058267 PCT/US2008/012224
(EntreMed, USA); pegaptanib (Pinn), (Gilead Sciences, USA); xanthorrhizol,
(Yonsei
University, South Korea); vaccine, gene-based, VEGF-2, (Scripps Clinic and
Research
Foundation, USA); SPV5.2, (Supratek, Canada); SDX 103, (University of
California at San
Diego, USA); PX 478, (Pro1X, USA); METASTATIN, (EntreMed, USA); troponin I,
(Harvard
University, USA); SU 6668, (SUGEN, USA); OXI 4503, (OXiGENE, USA); o-
guanidines,
Dimensional Pharmaceuticals, USA); motuporamine C, (British Columbia
University,
Canada); CDP 791, (Celltech Group, UK); atiprimod (p1NN), (GlaxoSmithKline,
UK); E 7820,
(Eisai, Japan); CYC 381, (Harvard University, USA); AE 941, (Aeterna, Canada);
vaccine,
angiogenesis, (EntreMed, USA); urokinase plasminogen activator inhibitor,
(Dendreon, USA);
oglufanide (pINN), (Melmotte, USA); HIF- lalfa inhibitors, (Xenova, UK); CEP
5214,
(Cephalon, USA); BAY RES 2622, (Bayer, Germany); Angiocidin, (InKine, USA);
A6,
(Angstrom, USA); KR 31372, (Korea Research Institute of Chemical Technology,
South
Korea); GW 2286, (GlaxoSmithKline, UK); EHT 0101, (ExonHit, France); CP
868596,
(Pfizer, USA); CP 564959, (OSI, USA); CP 547632, (Pfizer, USA); 786034,
(GlaxoSmithKline, UK); KRN 633, (Kirin Brewery, Japan); drug delivery system,
intraocular,
2-methoxyestradiol, (EntreMed, USA); anginex, (Maastricht University,
Netherlands, and
Minnesota University, USA); ABT 510, (Abbott, USA); AAL 993, (Novartis,
Switzerland);
VEGI, (ProteomTech, USA); tumor necrosis factor-alpha inhibitors, (National
Institute on
Aging, USA); SU 11248, (Pfizer, USA and SUGEN USA); ABT 518, (Abbott, USA);
YH16,
(Yantai Rongchang, China); S-3APG, (Boston Childrens Hospital, USA and
EntreMed, USA);
MAb, KDR, (ImClone Systems, USA); MAb, alpha5 betal, (Protein Design, USA);
KDR
kinase inhibitor, (Celltech Group, UK, and Johnson & Johnson, USA); GFB 116,
(South
Florida University, USA and Yale University, USA); CS 706, (Sankyo, Japan);
combretastatin
A4 prodrug, (Arizona State University, USA); chondroitinase AC, (IBEX,
Canada); BAY RES
2690, (Bayer, Germany); AGM 1470, (Harvard University, USA, Takeda, Japan, and
TAP,
USA); AG 13925, (Agouron, USA); Tetrathiomolybdate, (University of Michigan,
USA); GCS
100, (Wayne State University, USA) CV 247, (Ivy Medical, UK); CKD 732, (Chong
Kun
Dang, South Korea); MAb, vascular endothelium growth factor, (Xenova, UK);
irsogladine
(INN), (Nippon Shinyaku, Japan); RG 13577, (Aventis, France); WX 360, (Wilex,
Germany);
squalamine (p1NN), (Genaera, USA); RPI 4610, (Sirna, USA); cancer therapy,
(Marinova,
Australia); heparanase inhibitors, (InSight, Israel); KL 3106, (Kolon, South
Korea); Honokiol,
(Emory University, USA); ZK CDK, (Schering AG, Germany); ZK Angio, (Schering
AG,
Germany); ZK 229561, (Novartis, Switzerland, and Schering AG, Germany); XMP
300,
(XOMA, USA); VGA 1102, (Taisho, Japan); VEGF receptor modulators,
(Pharmacopeia,

33


CA 02703257 2010-04-20
WO 2009/058267 PCT/US2008/012224
USA); VE-cadherin-2 antagonists , (ImClone Systems, USA); Vasostatin,
(National Institutes
of Health, USA);vaccine, Flk-1, (ImClone Systems, USA); TZ 93, (Tsumura,
Japan);
TumStatin, (Beth Israel Hospital, USA); truncated soluble FLT I (vascular
endothelial
growth factor receptor 1), (Merck & Co, USA); Tie-2 ligands, (Regeneron, USA);
and,
thrombospondin I inhibitor, (Allegheny Health, Education and Research
Foundation, USA).
Alternatively, the present compounds may also be used in co-therapies with
other anti-
neoplastic agents, such as VEGF antagonists, other kinase inhibitors including
p38 inhibitors,
KDR inhibitors, EGF inhibitors (such as panitumumab), CDK inhibitors, TNF
inhibitors,
metallomatrix proteases inhibitors (MMP), COX-2 inhibitors including
celecoxib, NSAID's,

aõ133 inhibitors, phosphatidylinitisol 3-kinase inhibitors, AKT/PCK
inhibitors, proteasome
inhibitors (such as VelcadeTM), Trail receptor agonists (such as AMG 655),
Trail (such as
AMG 951), XIAP inhibitors, BCI2 inhibitors, Aurora kinase inhibitors, Raf
kinases inhibitors,
ubiquitin ligase inhibitors, HGF inhibitors (such as AMG 102), and c-Met
inhibitors (such as
compounds described WO 06/116713 and U.S. Serial No. 11/879,034).
Also included in the family of compounds of the current are the
pharmaceutically
acceptable salts and solvates thereof. The term "pharmaceutically-acceptable
salts" embraces
salts commonly used to form alkali metal salts and to form addition salts of
free acids or free
bases. The nature of the salt is not critical, provided that it is
pharmaceutically acceptable.
Suitable pharmaceutically acceptable acid addition salts of compounds of the
current invention
may be prepared from an inorganic acid or from an organic acid. Examples of
such inorganic
acids are hydrochloric, hydrobromic, hydroiodic, nitric, carbonic, sulfuric
and phosphoric acid.
Appropriate organic acids may be selected from aliphatic, cycloaliphatic,
aromatic,
arylaliphatic, heterocyclic, carboxylic and sulfonic classes of organic acids,
example of which
are formic, acetic, adipic, butyric, propionic, succinic, glycolic, gluconic,
lactic, malic, tartaric,
citric, ascorbic, glucuronic, maleic, fumaric, pyruvic, aspartic, glutamic,
benzoic, anthranilic,
mesylic, 4-hydroxybenzoic, phenylacetic, mandelic, embonic (pamoic),
methanesulfonic,
ethanesulfonic, ethanedisulfonic, benzenesulfonic, pantothenic, 2-
hydroxyethanesulfonic,
toluenesulfonic, sulfanilic, cyclohexylaminosulfonic, camphoric,
camphorsulfonic, digluconic,
cyclopentanepropionic, dodecylsulfonic, glucoheptanoic, glycerophosphonic,
heptanoic,
hexanoic, 2-hydroxy-ethanesulfonic, nicotinic, 2-naphthalenesulfonic, oxalic,
palmoic,
pectinic, persulfuric, 2-phenylpropionic, picric, pivalic propionic, succinic,
tartaric, thiocyanic,

mesylic, undecanoic, stearic, algenic, (3-hydroxybutyric, salicylic,
galactaric and galacturonic
acid. Suitable pharmaceutically-acceptable base addition salts of compounds of
the current
invention include metallic salts, such as salts made from aluminum, calcium,
lithium,

34


CA 02703257 2010-04-20
WO 2009/058267 PCT/US2008/012224
magnesium, potassium, sodium and zinc, or salts made from organic bases
including primary,
secondary and tertiary amines, substituted amines including cyclic amines,
such as caffeine,
arginine, diethylamine, N-ethyl piperidine, aistidine, glucamine,
isopropylamine, lysine,
morpholine, N-ethyl morpholine, piperazine, piperidine, triethylamine,
trimethylamine. All of
these salts may be prepared by conventional means from the corresponding
compound of the
invention by reacting, for example, the appropriate acid or base with the
compound of the
current invention. When a basic group and an acid group are present in the
same molecule, a
compound of the current invention may also form internal salts.
GENERAL SYNTHETIC PROCEDURES
The compounds of the invention can be synthesized according to the following
procedures.
The following abbreviations are used throughout the specification:
BH3 _ borane
EtOAc - ethyl acetate
HCl - hydrochloric acid
KOt-Bu - potassium butoxide
MeOH, CH3OH - methanol
NMP - N-methylpyrrolidinone, or (1-Methyl-2-pyrrolidinone)
K2CO3 - potassium carbonate
Tol - toluene

Example 1
H
O~ N /N\
N

O O
O N
7-((6,7-bis(methyloxy)-4-quinolinyl)oxy)-N-(5-methyl-3-isoxazolyl)-2,3-dihydro-
4H-1,4-
benzoxazine-4-carboxamide
The title compound was synthesized as described below.



CA 02703257 2010-04-20
WO 2009/058267 PCT/US2008/012224
0 H H
\ NHp CI CI O BH3 aN
HCI ~ N HCI
aq K2CO3, Tol MeOH/HCI HO / OH 75% HO p 95% HO
O
2 3
CI
MeO li~zz NMP, KOtBu
MeO N 100 C, 80%
N 4

p_NH
H
N \ N
O O / O
EtOAc / NMP
MeO cat. KOt-Bu MeO
,0
MeO \ ~N N MeO N

HN~ 5
O
0
b \ aq. K2CO3 / EtOac
/0
E N I
7 OY CI

\ O H2N 6
7-Hydroxy-2H-benzolbl I1,4Ioxazin-3(4H)-one (2)
K2CO3 (250g, 1.8 mot, 4 eq) was dissolved in water (1100 mL) in a 3 L round
bottomed flask,
vacuum degassed 3 times, then cooled to 0 to 5 C. 4-Amino resorcinol HCI (73
g, 0.45 mot, I
eq) was added though a strong nitrogen purge, and the blue solution was again
degassed 3x.
Choroacetyl chloride (45 mL, 64 g, 0.57 mot, 1.25 eq) was dissolved in toluene
(225 mL), and

was added over I h maintaining the temperature below 5 C. the ice bath was
removed, and the
mixture (biphasic with suspended crystalline product) was stirred overnight.
The intermediate
2 was then isolated via filtration giving 53g (71%) after a water wash (550
mL) and drying.
3,4-Dihydro-2H-benzolblll,41oxazin-7-ol hydrochloride (3)
To a 2 L round bottomed flask was charged with 7-hydroxy-2H-
benzo[b][1,4]oxazin-3(4H)-
one 2 (30.5 g, 185 mmol) and anhydrous THE (90 mL) under N2. The suspension
was cooled
36


CA 02703257 2010-04-20
WO 2009/058267 PCT/US2008/012224

to 5 C, and borane-THF complex (1 M in THF, 480 ml, 480 mmol, 2.6 equiv) was
added over
40 min under a stream of N2 to remove the H2 generated. Upon complete addition
(T=15 C),
the now colorless solution was removed from the ice bath and allowed to warm
to ambient
temperature. After stirring overnight, an aliquot was quenched into MeOH and
analyzed by

HPLC.

The solution was quenched by the slow addition (lh) of MeOH (80 mL) under a
stream of N2.
The mixture was then stirred at ambient temperature for 6 h to allow for
complete destruction
of the borate complex to product and trimethyl borane.
The resulting solution was concentrated by rotary evaporation, flushed with
methanol, and the
pale yellow residue was dissolved in MeOH (80 mL). The solution was then
cooled in an ice
bath under N2 and a solution of HCI (1 M in Et20, 200 mL, 200 mmol, 1.08 eq)
was added
over 10 min, giving the product as a white crystalline solid suspended in a
bright blue solution.
MTBE (120 ml-) was added over 30 min, and the mixture was allowed to age
overnight at
ambient temperature. The pale blue solid was filtered and washed nearly white
with
MTBE/MeOH (4: 1, 100 mL), then was dried under vacuum overnight to yield 32.7
g, (95%)
3 ,4-dihydro-2H-benzo[b] [ 1,4]oxazin-7-ol hydrochloride (3).
7-(6,7-dimethoxyguinolin-4-yloxy)-3,4-dihydro-2H-benzolbl11,41oxazine (5)
1-Methyl-2-pyrrolidinone (NMP), anhydrous 99.5% (12.6 L) was charged into a
100 L reactor
at ambient temperature followed by 3,4-dihydro-2H-benzo[b] [1, 4] oxazin-7-ol
hydrochloride
3 (1.5779 Kg, 8.41 mol, 1.5 equiv.) and 4-chloro-6, 7-dimethoxyquinoline 4
(1.2539 Kg, 5.61
mol, 1.0 equiv.) A 1.6 L portion of NMP was used to rinse the reactor walls.
Reactor contents
were cooled to 10 C. A series of 3 vacuum purges were performed on the
reactor. A charge of
potassium tert-butoxide, min 94.0% (1.9235 Kg, 17 mol, 3.0 equiv.) was
delivered into the
reactor in 3 portions.

A final 1.7 L portion of NMP was added as rinse. Reactor was purged with
vacuum/N2 cycles
3 more times. Reactor contents were heated to 100 C and subsequently held for
18 hours at an
agitation rate of 118 rpm. Completion was marked when 4-chloro-6,7-
dimethoxyquinoline was
<0.5% area at 220nm after 18 hours. The reactor contents were cooled to 25 C
and the reaction
mixture was seeded with 5.0 g of GMP penultimate, followed by purified water
addition of 9.3
L over a 10 minute period. The internal temperature rose by 30 C. The mixture
was cooled to
ambient and the remaining 22.5 L of purified water was charged in slowly. The
reactor

37


CA 02703257 2010-04-20
WO 2009/058267 PCT/US2008/012224
contents were held for 1 hour at 25 C with stir rate of 199 rpm. The thick off-
white slurry was
then transferred from the reactor through the centrifuge and into the
receiver. Filtration was
followed by the recirculation of filtrate over the wet cake to minimize the
product loss. In
addition, one NMP: water wash (2:1 ratio 10.5 L water: 5.25 L NMP) and one
water wash of
15.8 L was performed. Rotation speed on the centrifuge was 230 rpm for the
filtration and
water/NMP rinse. It was increased to 444 rpm for the water wash and was let to
spin out at 512
rpm for 2 hours.

Recrystallization of 5
A 100 L reactor was charged with 14.7 L of purified water and set to 20 C. The
wet cake was
transferred from the centrifuge in to the reactor followed by I -methyl-2-
pyrrolidinone (NMP),
anhydrous, 99.5% (7.35 L). The contents of the reactor were agitated at 135
rpm over 17 hours.
The thick off-white slurry was then transferred from the reactor through the
centrifuge and into
the receiver. Filtration was followed by the recirculation of filtrate over
the wet cake to
minimize the product loss. Reactor was rinsed and wet cake was washed with 45
L of purified
water in portions. Rotation speed on the centrifuge was 230 rpm for the
filtration, 340 rpm for
recirculation, 418 rpm for the first 15 L of water wash and 513 rpm for the
rest of the water
wash and spin out. Product was held spinning in the centrifuge under nitrogen
for 17 hours.
Subsequently, the batch was dried at 35 C in vacuum oven with high N2 flow for
7 days. The
process yielded 1.4008 Kg (73%) of 7-(6,7-dimethoxyquinolin-4-yloxy)-3,4-
dihydro-2H-
benzo[b] [ 1,4]oxazine. 5

Phenyl 5-methylisoxazol-3-y1carba mate (7)
Purified water, USP (9.5 L) was charged into a 100 L reactor at 20 C followed
by potassium
carbonate, (powder 325 mesh, 98+%, 3.3183 Kg, 24 mol, 2.5 equiv.); 3-amino-5-
methylisoxazole (942 g, 9.6 mol, 1.0 equiv.), and ethyl acetate (28.25 L, 30
volumes).
Additional 250 mL of water was used to rinse the walls of the reactor.
Contents of the reactor
were cooled to 5 C and 2 vacuum purges were performed. Phenyl chloroformate,
99%, (2.974
Kg, 19 mol, 2.0 equiv.) was charged slowly while maintaining the internal
temperature < 30 C.
At the end of addition the reactor temperature was set to 20 C with agitation
rate of 148 rpm.
Reactor contents were held for 90 minutes at 20 C resulting in a triphasic
mixture with solid
(product) in the mid layer. The reactor temperature was increased to 35 C in
order to dissolve
the solids and obtain a biphasic mixture more suitable for sampling.
Completion was marked
when 3-amino-5-methylisoxazole was <0.5% area at 220nm. The agitation was
stopped and the
layers were allowed to separate at 35 C. The aqueous layer was drained and a
10% sodium

38


CA 02703257 2010-04-20
WO 2009/058267 PCT/US2008/012224
carbonate solution (4.15 L) was charged and agitation resumed at 35 C. After
30 minutes the
agitation was stopped. The layers were separated and aqueous layer was
drained. The organic
layer was treated with 2 more purified water washes (6.5 L each) at 35 C.
Following the last
separation the organic layer was set for distillation with a jacket
temperature of 85 C. The
distillation was performed under a slight vacuum (12 psi) and a total of 18 L
of EtOAc was
collected.

The contents of the reactor were cooled to 0 C at ramp rate of 1.6 C / min and
held at 0 C for
2 hours. The thick white suspension was then transferred from the reactor
through the Aurora
filter and in to the receiver. Filtration was followed by the recirculation of
filtrate over the wet
cake and one cold EtOAc wash (2.0 L). The product was held on the filter under
nitrogen for 3
hours prior to the transfer to vacuum oven. The material was dried at 35 C
over 2 days with
strong nitrogen purge yielding 1.5450 Kg (74.3% yield) of 100% analytically
pure product. A
physical loss in the reactor of 96.23g, (4.6%) of 99.8% analytically pure
carbamate was later
observed. The loss in the ML was 375.8g, (18%) and in wash was 78.7g, (3.8%).

Title Compound
To a 100 L vessel was charged NMP (anhydrous, 1900 mL), 5 (1378 g, 4.07 mol,
100 mol%)
and ethyl acetate (12.5 L). The vessel was purged with nitrogen, agitation set
at 276 rpm, and
then the suspension was heated to 60 C. The solids dissolved, and the solution
was passed
though a 5 micron in-line filter (polypropylene) into a separate 100L vessel.
The filter was
rinsed with a 57 C mixture of NMP (185 mL) and EtOAc (1200 mL) prepared in the
initial
vessel.

To the initial vessel was charged NMP (1100 mL), 7 (1345 g, 6.16 mol, 151 mol
%) and
EtOAc (7100 L). The mixture was heated as above to 60 C to achieve
dissolution, and was
passed thought the same filter to the second vessel. A 60 C wash consisting of
NMP (185 mL)
and EtOAc (1200 mL) was passed though the filter.

To the second vessel was charged potassium t-butoxide (1.1 g), the vessel was
vacuum purged,
and the reaction was heated to 65 C. Within 1/2h, crystals had formed in the
reaction. After a
3h age, the reaction was assayed, and was deemed complete with 0.8 LCAP
penultimate
remaining.

39


CA 02703257 2010-04-20
WO 2009/058267 PCT/US2008/012224
The mixture was cooled to 25 C, and was aged overnight. The solid product was
collected by
filtration in an Aurora filter (5 micron polypropylene filter-cloth), and any
remaining product
left in the vessel was collected by liquor recycle. The product was washed
with EtOAc (4 x
2700 mL), and then was dried via nitrogen purge through the Aurora for 3 days.

A total of 1.7146 Kg (92%) of title compound was isolated as a light brown
solid. 'H NMR
(400 MHz, DMSO-d6) d ppm 10.16 (s, 1 H), 8.49 (d, J=5.31 Hz, 1 H), 7.70 (d,
J=8.84 Hz, I
H), 7.48 (s, I H), 7.39 (s, I H), 6.81 - 6.83 (m, 1 H), 6.76 - 6.80 (m, 1 H),
6.55 (d, J=5.18 Hz, I
H), 6.51 - 6.53 (m, I H), 4.27 - 4.31 (m, 2 H), 3.94 (s, 3 H), 3.93 (s, 3 H),
3.88 - 3.91 (m, 2 H),
2.37 (s, 3 H). HIZMS (M+H)+ calc'd: 463.1612 found: 463.1634.
Example 2

Compound A free base is known to exist in at least three anhydrous crystalline
forms as
well as a methanol solvate. All four forms have a unique powder X-ray powder
diffraction
(XRPD) patterns, the major peaks of which are shown below:

FORM Powder X-ray Diffraction Peaks (Approximate 2Theta)
Form I 4.5, 9.5, 16.0, 24.0

Form II 5.6, 8.8, 11.2, 13.6, 14.9, 24.8
Form III 10.3, 13.5, 14.3, 16.4, 19.6
Methanol Solvate 5.7, 8.9, 9.3, 11.3, 11.9, 13.1, 18.4, 24.8, 26.7

With heating at 10 C/minute, anhydrous Form I converts to anhydrous Form II
at a
temperature of -150 C, and Form II decomposes near 240 C. Form III
decomposes near 235
C. The methanol solvate liberates methanol near 135 C and converts to
anhydrous Form II.
Compound A has also been shown to form crystalline salts with methanesulfonic
acid,
phosphoric acid, sulfuric acid and hydrochloric acid. The mesylate salt is
known to exist in at
least two crystalline forms with unique X-ray powder diffraction patterns, the
major peaks of
which are shown below:

FORM Powder X-ray Diffraction Peaks (Approximate 2Theta)
Form I 6.7, 10.2, 12.0, 14.9, 17.8

Form II 4.2, 4.9, 9.9, 22.1, 25.6


CA 02703257 2010-04-20
WO 2009/058267 PCT/US2008/012224
Form I mesylate salt has been formed by suspending the free base in EtOH,
adding a
slight molar excess of methanesulfonic acid, heating to dissolve and slowly
cooling to room
temperature. The resulting solid was then isolated by filtration. Form II
mesylate salt has been
formed by suspending the free base in isopropyl alcohol (IPA), adding a slight
molar excess of
methanesulfonic acid, heating to dissolve and slowly cooling to room
temperature. The
resulting solid was then isolated by filtration.

The hydrochloride salt is formed by preparing a solution of Compound A free
base in
ethanol and adding hydrochloric acid in slight molar excess with stirring and
optional heat.
Spontaneous salt formation occurred both with and without the application of
heating and
cooling. The hydrochloride salt is known to exist in at least one anhydrous
form, which has
been confirmed by single-crystal X-ray analysis, and at least one hydrous
form. The
anhydrous form of the hydrochloride salt is crystalline and decomposes near
230 C when
heated at a rate of 10 C/minute. The hydrous form of Compound A hydrochloride
has been
obtained by suspending the anhydrous hydrochloride material in water or in
dilute aqueous
solutions of hydrochloric acid. The hydrous material is crystalline, can
freely absorb up to 4
molar equivalents of water at 90% relative humidity and 25 C, and appears to
fully dehydrate
by 150 C and decompose near 170 C when heated at a rate of 10 C/minute.
Both the
anhydrous and hydrous forms of Compound A hydrochloride have unique X-ray
powder
diffraction patterns, the major peaks of which are described below:

FORM Powder X-ray Diffraction Peaks (Approximate 2Theta)
Anhydrous 5.2, 10.5, 12.0, 15.5, 17.3, 19.7, 24.2
Hydrous 8.4, 9.5, 11.2, 12.3, 16.9, 24.5

Apparent phosphoric acid salt material was generated by high throughput
screening out
of one or more of the following: Acetone: Water (1:1), Tetrahydrofuran (THF),
THF:Water
(9:1), methyl ethyl ketone (MEK), isopropyl acetate (IPAc), toluene, ethanol,
ethanol:water
(9:1), isopropyl alcohol (IPA), IPA:water (9:1) and acetonitrile. Samples were
initially
prepared by: 1) adding free base to the crystallization source plate, 2)
adding a slight molar

41


CA 02703257 2012-03-13

WO 2009/058267 PCT/US2008/012224
excess of acid, 3) adding the crystallization solvent to a final concentration
of -S 16 mg/mL, and
4) heating to 55 C for 1 hour with stirring. The samples were then filtered
and split for
crystallization by evaporation, cooling or anti-solvent addition with n-butyl
ether. The
crystallization plates were centrifuged, and the supernatants were aspirated
allowing for
microscopic and XRPD analyses of any resulting solids. Any solid remaining in
the source
plate was also analyzed by microscopy and XRPD. The experiments with
phosphoric acid
resulted in two possible crystalline salts.

Apparent sulfuric acid salt material was generated by high throughput
screening as
described above. The experiments with sulfuric acid resulted in two possible
crystalline salts.
Apparent hydrochloric acid salt material was generated by high throughput
screening as
described above. The high throughput experiments with hydrochloric acid
resulted in two
possible crystalline salts.
Apparent methanesulfonic acid salt material was generated by high throughput
screening as described above. The high throughput experiments with
methanesulfonic acid
resulted in three possible crystalline salts.

Of course, it will be understood that salts can be generated using different
organic
solvents than those described above. It is believed that other acid addition
salts of Compound
A are possible with counter-ions from organic and inorganic acids, preferably
those having
pKa < 5. It is also understood that additional physical forms of free base and
various salts may
exist.

Example 3
In Vitro Metabolite Profiling
incubation mixtures were prepared including [14C]-Compound A (10 M, 0.5 MCi),
liver
microsomes at I mg/mL (human, mouse, rat, monkey, rabbit, dog), MgC12.(3 mM),
and
potassium phosphate buffer (100 mM). The reaction was started by adding NADPH
(1 mM)
and incubations were carried out in a shaking water bath maintained at 37 C
for 1 hr. The
reactions were stopped by adding I volume of ACN:MeOH 1:1 mixture; vortex
mixed;
centrifuged at 16000 xg. The supernatants were analyzed on a reverse phase
HPLC (Agilent*
1100, Agilent systems, DE) in line with a radiomatic ([3-ram, IN/US systems,
FL) detector and

42
*Trademark


CA 02703257 2010-04-20
WO 2009/058267 PCT/US2008/012224
an LTQ ion-trap mass spectrometer (ThermoFisher, San Jose, CA). HPLC
separations were
carried out on a phenyl-hexyl luna (150 x 4.6 mm, 5 m, Phenomenex Inc) column
maintained
at 40 C at a flow rate of I mL/min. A binary mobile phase consisting of 10 mM
ammonium
formate in H,O (solvent A) and 0.1 % formic in MeOH (solvent B) was employed
under the
following gradient conditions: 0 to 2 min, 95% A; 2 to 10 min, 95 to 75%A; 10
to 40 min, 75
to 60% A; 40 to 60 min, 60 to 55 % A; 60 to 77 min, 55 to 25%A; 77 to 78 min,
25 to 5% A;
78 to 83 min, 5% A; 83 to 84 min, 5 to 95%A; 84 to 90 min, 95%A. The flow post-
column was
split such that 80% went to radiomatic detector and the remaining 20% to MS.
The flow into
radiomatic detector was mixed with 3 volumes of liquid scintillant
(UltimaFlow, PerkinElmer)
prior to radio-detection.

Substantial turnover of Compound A was observed in all species. The following
compounds
identified in Table 16 below were the significant metabolites formed in all
species. These same
metabolites were also formed upon incubating Compound A with recombinant human
cytochrome P450 3A4 isomform fortified with NADPH.
TABLE 16

Metabolite KDR Retention NMR
Inhibition Time
(ILM) (min)
i 1 1 H NMR (400 MHz,
N DMSO-d6) ^ ppm 10.15
(s, 1 H), 8.31 (d, J=6.82
0\ NH Hz, I H), 7.93 (s, I H),
DIY 5 75 7.68 (d, J=8.84 Hz, I
H), 7.47 (s, I H), 6.74 -
Me I ) 6.86 (m, 2 H), 6.58 (d,
J=6.95 Hz, I H), 6.52 (s,
I H),4.25-4.31 (m,2
0 N H), 3.99 (s, 3 H), 3.95
Me o (s, 3 H), 3.85 - 3.91 (m,
2 H), 2.37 (s, 3 H).

43


CA 02703257 2010-04-20
WO 2009/058267 PCT/US2008/012224
Metabolite KDR Retention NMR
Inhibition Time
(M min
1 H NMR (400 MHz,
N DMSO-d6) ^ ppm 10.16
(s, I H), 10.10 (s, I H),
y NH 0.777202 59.5 8.42 (d, J=5.2 Hz, 1 H),
7.69 (d, J=9.0 Hz, I H),
7.45 (s, I H), 7.27 (s, I
Me H), 6.73 - 6.83 (m, 2 H),
1 6.52 (s, I H), 6.47 (d,
)a,"- J=5.3 Hz, I H), 4.24 -
4.32 (m, 2 H), 3.93 (s, 3
HO H), 3.84 - 3.92 (m, 2 H),
2.37 (s, 3 H)

0 1 H NMR (400 MHz,
Ni 1 DMSO-d6) ^ ppm 10.06
(s, 2 H), 8.44 (d, J=5.2
0`Z Y NH Hz, I H), 7.67 (d, J=8.8
0.001684 55.1 Hz, I H), 7.42 (s, I H),
7.36 (s, I H), 6.68 - 6.80
(m, 2 H), 6.48 - 6.57 (m,
xx: 2 H), 4.23 - 4.33 (m, 2
HO H), 3.94 (s, 3 H), 3.83 -
3.91 (m, 2 H), 2.37 (s, 3
H)
Me N

0 off 1 H NMR (400 MHz,
DMSO-d6) ^ ppm 10.29
(s, 1 H), 8.81 (d, J=6.57
o\ /NH Hz, I H), 7.81 (d,
DIY 0.001042 55.1 J=8.97 Hz, I H), 7.71 (s,
"~ 1 H), 7.57 (s, I H), 6.98
\ - 7.02 (m, I H), 6.89 -
1 e 6.94 (m, 2 H), 6.67 (s, 1
H), 4.53 (s, 2 H), 4.30 -
4.36 (m, 2 H), 3.99 -
N
1 4.06 (m, 6 H), 3.89 -
Me 3.95 (m, 2 H).
Identification of the chemical structure of the metabolites was achieved
through
comparison with synthesized standards. The Compound A N-oxide standard was
synthesized
using procedures known in the art. The remaining metabolites was sysnthesized
as follows:
44


CA 02703257 2010-04-20
WO 2009/058267 PCT/US2008/012224
N
O\ _NH

/ N
lMe O I O
~

HO \

Metabolite

H
CI
N Cs2CO3, DMF O O
Me0 /
80 ,C MeO
BnO N HO \ O
HCI
BnO \ N
7-(7-(benzyloxy)-6-methoxyquinolin-4-yloxy)-3,4-dihydro-2H-
benzo[bI11,41oxazine
A solution of 3,4-dihydro-2H-benzo[b][1,4]oxazin-7-ol hydrochloride (0.626 g,
3.34 mmol),
cesium carbonate (4.35 g, 13.3 mmol), and 7-(benzyloxy)-4-chloro-6-
methoxyquinoline (1.000
g, 3.34 mmol) in DMF (10.0 mL) was stirred at 80 C. After 17 h, additional
3,4-dihydro-2H-
benzo[b][1,4]oxazin-7-ol hydrochloride (0.626 g, 3.34 mmol) was added, and the
mixture
stirred at 80 C for 20 h. Additional 3,4-dihydro-2H-benzo[b][1,4]oxazin-7-ol
hydrochloride
(0.6259 g, 3.336 mmol) and cesium carbonate (4.35 g, 13.3 mmol) were added,
and the
mixture stirred at 80 C for 3 days. The reaction mixture was partitioned
between
dichloromethane and water. The aqueous phase was separated and extracted with
dichloromethane. The combined organic phases were washed with brine, dried
over anhydrous
sodium sulfate, filtered, and concentrated to afford an orange-brown oil. This
material was
purified twice via column chromatography (RediSep 120 g column, gradient
elution with 0-5%
methanol-dichloromethane) to afford an orange oil. Trituration with methanol
and filtering the
resulting suspension afforded 7-(7-(benzyloxy)-6-methoxyquinolin-4-yloxy)-3,4-
dihydro-2H-
benzo[b][1,4]oxazine (0.560 g, 40.5% yield) as a tan solid. MS (MH+) 415.1;
Calculated 414
for C25H22N204.



CA 02703257 2012-03-13

WO 2009/058267 PCT/US2008/012224
H H
N / N
Hp, PdWC \ I Jl
O IO O O
00
Me0 / EtOH, rt MeO

PhO N HO \ N
4-(3,4-dihydro-2H-benzoIbi 11,41oxazin-7-yloxy)-6-methoxyquinolin-7-ol
Palladium, 10 wt% on activated carbon (0.050 g, 0.47 mmol) was added to a
solution of 7-(7-
(benzyloxy)-6-methoxyquinolin-4-yloxy)-3,4-dihydro-2H-benzo[b][I,4]oxazine
(0.250 g, 0.60
mmol) in ethanol (10.0 mL). The system was evacuated and purged with hydrogen
(g) three
times and then stirred under a H2 (g) atmosphere for 20 h. The reaction
mixture was filtered
through a pad of Celite and concentrated to afford 4-(3,4-dihydro-2H-
benzo[b][1,4]oxazin-7-
yloxy)-6-methoxyquinolin-7-ol (0.200 g, 102% yield) as a yellow-green solid.
MS (MH+)
325.0; Calculated 324 for Cj8H16N204,

_N
O
N1 _N OYNH
O \ I pJ o o NH N
Et3N. THE
-a 1
MeO \ / O 50 e0 O \ OJ
' I MeO
HO N OiN -i
`
HO N
7-(7-hydroxy-6-methoxyquinolin-4-yloxy)-N-(5-methylisoxazol-3-yl)-2,3-
dihydrobenzolbff 1,4]oxazine-4-carboxamide
A 25-mL round bottomed flask was charged with 4-(3,4-dihydro-2H-
benzo[b][1,4]oxazin-7-
yloxy)-6-methoxyquinolin-7-ol (0.124 g, 0.38 mmol), 4-nitrophenyl 5-
methylisoxazol-3-
ylcarbamate (0.11 g, 0.40 mmol), and THE (5.0 mL). Triethylamine (0.160 mL,
1.1 mmol)
was added, the system was purged with argon, and the tube was sealed. The
mixture stirred at
50 C for 16 h. The reaction mixture was concentrated and the residue was
purified via
column chromatography on silica gel (RediSep 40 g column, 100% ethyl acetate
for 10 min,
followed by 5% methanol/dichloromethane for 20 min) to afford 7-(7-hydroxy-6-
methoxyquinolin-4-yloxy)-N-(5-methylisoxazol-3-yl)-2,3-dihydrobenzo[b][l
,4]oxazine-4-
carboxamide (0.026 g, 15% yield) as a yellow solid.

*Trademark

46


CA 02703257 2010-04-20
WO 2009/058267 PCT/US2008/012224
N
O\ NH

N

0 0
HO /

I \ N/
Me

Metabolite
H
N
CI H
BnO N Cs2CO3, DMF 0 0
80-C BnO /
MeO \ N HO \ O
HCI MeO \ N

7-(6-(benzyloxy)-7-methoxyquinolin-4-yloxy)-3,4-dihydro-2H-
benzoIbil1,41oxazine
A solution of 3,4-dihydro-2H-benzo[b][1,4]oxazin-7-ol hydrochloride (0.626 g,
3.34 mmol),
cesium carbonate (4.35 g, 13.3 mmol), and 6-(benzyloxy)-4-chloro-7-
methoxyquinoline (1.00
g, 3.34 mmol) in DMF (10.0 mL) was stirred at 80 C for 18 h. An additional
3,4-dihydro-2H-
benzo[b][1,4]oxazin-7-ol hydrochloride (0.626 g, 3.34 mmol) and cesium
carbonate (4.35 g,
13.3 mmol) were added, and the mixture stirred at 80 C for 3 days. The
reaction mixture was
partitioned between dichloromethane and water. The aqueous phase was separated
and
extracted with dichloromethane. The combined organic phases were washed with
brine, dried
over anhydrous sodium sulfate, filtered, and concentrated to afford an orange-
brown oil. This
material was purified twice via column chromatography (RediSep 80 g column,
gradient
elution with 0-5% methanol-dichloromethane) to afford an orange oil.
Trituration with
methanol and filtering the resulting suspension afforded 7-(6-(benzyloxy)-7-
methoxyquinolin-
4-yloxy)-3,4-dihydro-2H-benzo[b][1,4]oxazine (1.156 g, 83.6% yield) as an
orange glass. MS
(MH+) 415.1; Calculated 414 for C25H22N204=


47


CA 02703257 2010-04-20
WO 2009/058267 PCT/US2008/012224
/ N / N
\ I H2, Pd/C \
O O O O
Ph 1--l-10 / EtOH, rt
HO / \
MeO \ N\ MeO N
4-(3,4-dihydro-2H-benzo[b] [1,4]oxazin-7-yloxy)-7-methoxyquinolin-6-ol
Palladium, 10 wt% on activated carbon (0.100 g, 0.940 mmol) was added to a
solution of 7-(6-
(benzyloxy)-7-methoxyquinolin-4-yloxy)-3,4-dihydro-2H-benzo[b][1,4]oxazine
(1.156 g, 2.79
mmol) in ethanol (20.0 mL). The system was evacuated and purged with hydrogen
(g) three
times and then stirred under a H2 (g) atmosphere for 18 h. Additional
palladium, 10 wt% on
activated carbon (0.100 g, 0.940 mmol) was added to the reaction mixture. The
system was
evacuated and purged with hydrogen (g) three times and then stirred under a H2
(g) atmosphere
for 20 h. The reaction mixture was filtered through a pad of Celite and
concentrated to afford
an orange brown solid. This material was purified via column chromatography on
silica gel
(RediSep 40 g column, gradient elution with 0-100% (90:10:1,
dichloromethane/methanol/ammonium hydroxide)-dichloromethane) to afford 4-(3,4-
dihydro-
2H-benzo[b][1,4]oxazin-7-yloxy)-7-methoxyquinolin-6-ol (0.733 g, 81.0% yield)
as a yellow
solid. MS (MH+) 325.0; Calculated 324 for C18H16N204.

O
H N / O N
\ I ~ N 0 NH
Et3N, THE Y
O O O NH N/
HO \ 50 C
O
~ O \ O
MeO \ N
02N \ HO / \
MeO N

7-(6-hydroxy-7-methoxyquinolin-4-yloxy)-N-(5-methylisoxazol-3-yl)-2,3-
dihydrobenzo[ b[ [ 1,41oxazine-4-carboxamide
A resealable tube was charged with 4-(3,4-dihydro-2H-benzo[b] [ 1,4]oxazin-7-
yloxy)-7-
methoxyquinolin-6-ol (0.200 g, 0.617 mmol), 4-nitrophenyl 5-methylisoxazol-3-
ylcarbamate
(0.243 g, 0.925 mmol), and THE (5.0 mL). Triethylamine (0.258 mL, 1.85 mmol)
was added,
the system was purged with argon, and the tube was sealed. The mixture stirred
at 50 C for
15 h. The reaction mixture was partitioned between ethyl acetate and water.
The aqueous
phase was separated and extracted with ethyl acetate. The combined organic
phases were
48


CA 02703257 2010-04-20
WO 2009/058267 PCT/US2008/012224
washed with water and brine, dried over anhydrous sodium sulfate, filtered,
and concentrated
to afford a yellow-orange oil. This material was purified via column
chromatography on silica
gel (RediSep 40 g column, eluting with 5% methanol-dichloromethane) to afford
a yellow oil.
This material was triturated with dichloromethane and filtered to afford 7-(6-
hydroxy-7-
methoxyquinolin-4-yloxy)-N-(5-methylisoxazol-3-yl)-2,3-dihydrobenzo[b] [
1,4]oxazine-4-
carboxamide (0.104 g, 38% yield) as an off-white solid.

OH
N

O~y NH
O N
Me O
N
Me

Metabolite
A.

Et3N, OH TBSCI,
HO.N N- 0 OH Et N O OTBS
N 3 I
Il ~ /
Et02C CI CH2CI2 Et02C DMF EtO2C
1 2
N,0 OTBS N,p OH
H2NNH2-H20 O NaNO2 0
EtOH HCI
H2N_ NH N3
3 4

A solution of ethyl chlorooximidoacetate (15 g, 0.1 mol) in CH2C12 is added
dropwise
over 4 hours to propargyl alcohol (29 mL, 0.5 mol) and Et3N (14 mL, 0.1 mmol)
in 200 mL
CH2C12. When the addition is complete the reaction mixture is concentrated and
triturated with
Et20. The solid is filtered and the organics are concentrated again. The
remaining oil is
chromatographed over silica gel (EtOAc/Hex: 20/80) to give 5-
(hydroxymethyl)isoxazole-3-

49


CA 02703257 2010-04-20
WO 2009/058267 PCT/US2008/012224
carboxylate as an oil. The remaining propargyl alcohol is removed by
azeotroping from n-
heptane.
To a solution of 5-(hydroxymethyl)isoxazole-3-carboxylate (4.0 g, 23 mmol) and
TBSCL (3.7 g, 25 mmol) in DMF is added Et3N (3.4 mL, 24 mmol) dropwise over 20
minutes.
The reaction is allowed to stir for 30 minutes after which it is diluted with
EtOAc (300 mL),
washed with I M HCl (3 x 100 mL), 5% CuSO4 (2 x 50 mL) and concentrated in
vacuo to
yield ethyl-5-({[tert-butyl(dimethylsilyl)]oxy}methyl)isoxazole-3-carboxylate
as an oily
material.
A mixture of ethyl -5-({[tert-butyl(dimethylsilyl)]oxy} methyl)isoxazole-3-
carboxylate
(1.68 g, 5.9 mmol) and hydrazine hydrate (44 g, 8.8 mmol) in ethanol (30 mL)
is heated to
60 C for 4 hours. The mixture is cooled to room temperature and the solvents
are removed in
vacuo to yield 5-({[tert-butyl(dimethyl)silyl]oxy}methyl)isoxazole-3-
carbohydrazide as orange
crystals.
A mixture of 5-({[tent-butyl(dimethyl)silyl]oxy}methyl)isoxazole-3-
carbohydrazide
(1.32 g, 4.9 mmol) in concentrated HCl (40 mL) is cooled to 0 C, followed by a
dropwise
addition of aqueous NaNO2 (0.42 g, 6.1 mL), maintaining the temperature below
5 C. After I
hour, the mixture is diluted with water (100 mL) and extracted with EtOAc (3 x
50 mL).
Organics are dried (MgSO4) and concentrated in vacuo to yield azido(5-
(hydroxymethyl)isoxazol-3-yl)methanone (4) as tan crystals.

B.
N,0 OH TBDMSCI, N,0 OTBDMS
l X Imidazole
O ~ O
CH2C12
N3 N3
4 5
Azido(5-((tert-butyldimethylsilyloxy)methyl)isoxazol-3-yl)methanone (5).
To a solution of azido(5-(hydroxymethyl)isoxazol-3-yl)methanone (4) (100 mg,
0.60
mmol) and tert-butyldim ethyl sil yl chl ori de (99 mg, 0.65 mmol) in CH2C12
(3 mL) at 0 C was
added imidazole (49 mg, 0.71 mmol). After 4 hrs, the mixture was allowed to
warm to RT and
stirred for an additional 14 hrs. The mixture was cooled to 0 C and filtered
through a short
silica gel plug. The plug was rinsed with cold CH2C12 and the filtrate was
concentrated in
vacuo to afford crude azido(5-((tert-butyldimethylsilyloxy)methyl)isoxazol-3-
yl)methanone
(150 mg, 89% yield) as a white solid that was advanced without further
purification. MH+ _
283.1.



CA 02703257 2010-04-20
WO 2009/058267 PCT/US2008/012224
C.
OTBDMS
0
H N-0 OTBDMS N
N
O 0 NH
O 0 N3 () Y
O )C e-N I THE I ~ O O / O

6
0 \ -N
7
N-(5-((tcrt-butyldimethylsilyloxy)methyl)isoxazol-3-yl)-7-(6,7-
dimethoxyquinolin-4-
yloxy)-2,3-dihydrobenzo[b1[1,4]oxazine-4-carboxamide (7).
To a resealable tube was added azido(5-((tert-
butyldimethylsilyloxy)methyl)isoxazol-
3-yl)methanone (5) (50 mg, 0.18 mmol), 7-(6,7-dimethoxyquinolin-4-yloxy)-3,4-
dihydro-2H-
benzo[b] [ 1,4]oxazine (6) (20 mg, 0.059 mmol) and THE (1 mL). The tube was
sealed and
heated to 80 C for 4 hrs. The solvent was removed in vacuo and the residue
was purified by
silica gel chromatography using 50-100% Hexanes:EtOAc to afford N-(5-((tert-
butyldimethylsilyloxy)methyl)isoxazol-3-yl)-7-(6,7-dimethoxyquinolin-4-yloxy)-
2,3-
dihydrobenzo[b][1,4]oxazine-4-carboxamide (27 mg, 75% yield) as a white solid.
MH+ _
593.2.
D.
OTBDMS OH
O O
ONH HCI O`~NH

\ N EtOH \ N
c
O O O o
HCI
0 \ \N O \ \N
7 8
7-(6,7-dimethoxyquinolin-4-yloxy)-N-(5-(hydroxymethyl)isoxazol-3-yl)-2,3-
dihydrobenzo[b1[1,4]oxazine-4-carboxamide hydrochloride (8).
To N-(5-((tert-butyldimethylsilyloxy)methyl)isoxazol-3-yl)-7-(6,7-
dimethoxyquinolin-
4-yloxy)-2,3-dihydrobenzo[b][1,4]oxazine-4-carboxamide (7) (50 mg, 0.084 mmol)
was added
51


CA 02703257 2010-04-20
WO 2009/058267 PCT/US2008/012224

2 N HCI in EtOH (5 mL) at RT. After 15 min, the solid had dissolved and the
solution was
stirred at RT for an additional 2 hrs at which time a white precipitate
formed. The mixture was
cooled to 0 C, filtered, and the isolated solid was washed with cold EtOH.
The solid was
dried in vacuo to afford 7-(6,7-dimethoxyquinolin-4-yloxy)-N-(5-
(hydroxymethyl)isoxazol-3-
yl)-2,3-dihydrobenzo[b][1,4]oxazine-4-carboxamide hydrochloride (33 mg, 83%
yield). MH+
= 479.2.

FORMULATIONS
Also embraced within this invention is a class of pharmaceutical compositions
comprising the active compounds of the current invention in association with
one or more non-
toxic, pharmaceutically-acceptable carriers and/or diluents and/or adjuvants
(collectively
referred to herein as "carrier" materials) and, if desired, other active
ingredients. The active
compounds of the present invention may be administered by any suitable route,
preferably in
the form of a pharmaceutical composition adapted to such a route, and in a
dose effective for
the treatment intended. The compounds and compositions of the present
invention may, for
example, be administered orally, mucosally, topically, rectally, pulmonarily
such as by
inhalation spray, or parentally including intravascularly, intravenously,
intraperitoneally,
subcutaneously, intramuscularly intrasternally and infusion techniques, in
dosage unit
formulations containing conventional pharmaceutically acceptable carriers,
adjuvants, and
vehicles.
The pharmaceutically active compounds of this invention can be processed in
accordance with conventional methods of pharmacy to produce medicinal agents
for
administration to patients, including humans and other mammals.
For oral administration, the pharmaceutical composition may be in the form of,
for
example, a tablet, capsule, suspension or liquid. The pharmaceutical
composition is preferably
made in the form of a dosage unit containing a particular amount of the active
ingredient.
Examples of such dosage units are tablets or capsules. For example, these may
contain an
amount of active ingredient from about 50 micrograms to 2000 mg, preferably
from about I to
500 mg. A suitable daily dose for a human or other mammal may vary widely
depending on
the condition of the patient and other factors, but, once again, can be
determined using routine
methods.
The amount of compounds which are administered and the dosage regimen for
treating
a disease condition with the compounds and/or compositions of this invention
depends on a
variety of factors, including the age, weight, sex and medical condition of
the subject, the type
52


CA 02703257 2010-04-20
WO 2009/058267 PCT/US2008/012224
of disease, the severity of the disease, the route and frequency of
administration, and the
particular compound employed. Thus, the dosage regimen may vary widely, but
can be
determined routinely using standard methods. A daily dose of about 0.001 to
500 mg/kg,
preferably between about 0.01 and about 50 mg/kg, and more preferably about
0.001 and about
30 mg/kg body weight may be appropriate. The daily dose can be administered in
one to four
doses per day.
For therapeutic purposes, the active compounds of this invention are
ordinarily
combined with one or more adjuvants appropriate to the indicated route of
administration. If
administered per os, the compounds may be admixed with lactose, sucrose,
starch powder,
cellulose esters of alkanoic acids, cellulose alkyl esters, talc, stearic
acid, magnesium stearate,
magnesium oxide, sodium and calcium salts of phosphoric and sulfuric acids,
gelatin, acacia
gum, sodium alginate, polyvinylpyrrolidone, and/or polyvinyl alcohol, and then
tableted or
encapsulated for convenient administration. Such capsules or tablets may
contain a controlled-
release formulation as may be provided in a dispersion of active compound in
hydroxypropylmethyl cellulose.
In the case of psoriasis and other skin conditions, it may be preferable to
apply a topical
preparation of compounds of this invention to the affected area two to four
times a day.
Formulations suitable for topical administration include liquid or semi-liquid
preparations suitable for penetration through the skin (e.g., liniments,
lotions, ointments,
creams, or pastes) and drops suitable for administration to the eye, ear, or
nose. A suitable
topical dose of active ingredient of a compound of the invention is 0.1 mg to
150 mg
administered one to four, preferably one or two times daily. For topical
administration, the
active ingredient may comprise from 0.001 % to 10% w/w, e.g., from I% to 2% by
weight of
the formulation, although it may comprise as much as 10% w/w, but preferably
not more than
5% w/w, and more preferably from 0.1 % to I% of the formulation.
When formulated in an ointment, the active ingredients may be employed with
either
paraffinic or a water-miscible ointment base. Alternatively, the active
ingredients may be
formulated in a cream with an oil-in-water cream base. If desired, the aqueous
phase of the
cream base may include, for example at least 30% w/w of a polyhydric alcohol
such as
propylene glycol, butane-1,3-diol, mannitol, sorbitol, glycerol, polyethylene
glycol and
mixtures thereof. The topical formulation may desirably include a compound,
which enhances
absorption or penetration of the active ingredient through the skin or other
affected areas.
Examples of such dermal penetration enhancers include DMSO and related
analogs.

53


CA 02703257 2012-03-13

WO 2009/058267 PCT/US2008/012224
The compounds of this invention can also be administered by a transdermal
device.
Preferably transdermal administration will be accomplished using a patch
either of the
reservoir and porous membrane type or of a solid matrix variety. In either
case, the active agent
is delivered continuously from the reservoir or microcapsules through a
membrane into the
active agent permeable adhesive, which is in contact with the skin or mucosa
of the recipient.
If the active agent is absorbed through the skin, a controlled and
predetermined flow of the
active agent is administered to the recipient. In the case of microcapsules,
the encapsulating
agent may also function as the membrane.
The oily phase of the emulsions of this invention may be constituted from
known
ingredients in a known manner. While the phase may comprise merely an
emulsifier, it may
comprise a mixture of at least one emulsifier with a fat or an oil or with
both a fat and an oil.
Preferably, a hydrophilic emulsifier is included together with a lipophilic
emulsifier, which
acts as a stabilizer. It is also preferred to include both an oil and a fat.
Together, the
emulsifier(s) with or without stabilizer(s) make-up the so-called emulsifying
wax, and the wax
together with the oil and fat make up the so-called emulsifying ointment base,
which forms the
oily dispersed phase of the cream formulations. Emulsifiers and emulsion
stabilizers suitable
for use in the formulation of the present invention include Tween 60, Span 80,
cetostearyl
alcohol, myristyl alcohol, glyceryl monostearate, sodium lauryl sulfate,
glyceryl distearate
alone or with a wax, or other materials well known in the art.
The choice of suitable oils or fats for the formulation is based on achieving
the desired
cosmetic properties, since the solubility of the active compound in most oils
likely to be used
in pharmaceutical emulsion formulations is very low. Thus, the cream should
preferably be a
non-greasy, non-staining and washable product with suitable consistency to
avoid leakage from
tubes or other containers. Straight or branched chain, mono- or dibasic alkyl
esters such as di-
isoadipate, isocetyl stearate, propylene glycol diester of coconut fatty
acids, isopropyl
myristate, decyl oleate, isopropyl palmitate, butyl stearate, 2-ethylhexyl
palmitate or a blend of
branched chain esters may be used. These may be used alone or in combination
depending on
the properties required. Alternatively, high melting point lipids such as
white soft paraffin
and/or liquid paraffin or other mineral oils can be used.
Formulations suitable for topical administration to the eye also include eye
drops
wherein the active ingredients are dissolved or suspended in suitable carrier,
especially an
aqueous solvent for the active ingredients. The active ingredients are
preferably present in
such formulations in a concentration of 0.5 to 20%, advantageously 0.5 to 10%
and particularly
about 1.5% w/w.

*Trademark 54


CA 02703257 2010-04-20
WO 2009/058267 PCT/US2008/012224
Formulations for parenteral administration may be in the form of aqueous or
non-
aqueous isotonic sterile injection solutions or suspensions. These solutions
and suspensions
may be prepared from sterile powders or granules using one or more of the
carriers or diluents
mentioned for use in the formulations for oral administration or by using
other suitable
dispersing or wetting agents and suspending agents. The compounds may be
dissolved in
water, polyethylene glycol, propylene glycol, ethanol, corn oil, cottonseed
oil, peanut oil,
sesame oil, benzyl alcohol, sodium chloride, tragacanth gum, and/or various
buffers. Other
adjuvants and modes of administration are well and widely known in the
pharmaceutical art.
The active ingredient may also be administered by injection as a composition
with suitable
carriers including saline, dextrose, or water, or with cyclodextrin (ie.
Captisol), cosolvent
solubilization (ie. propylene glycol) or micellar solubilization (ie. Tween
80).
The sterile injectable preparation may also be a sterile injectable solution
or suspension
in a non-toxic parenterally acceptable diluent or solvent, for example as a
solution in 1,3-
butanediol. Among the acceptable vehicles and solvents that may be employed
are water,
Ringer's solution, and isotonic sodium chloride solution. In addition,
sterile, fixed oils are
conventionally employed as a solvent or suspending medium. For this purpose
any bland fixed
oil may be employed, including synthetic mono- or diglycerides. In addition,
fatty acids such
as oleic acid find use in the preparation of injectables.
For pulmonary administration, the pharmaceutical composition may be
administered in
the form of an aerosol or with an inhaler including dry powder aerosol.
Suppositories for rectal administration of the drug can be prepared by mixing
the drug
with a suitable non-irritating excipient such as cocoa butter and polyethylene
glycols that are
solid at ordinary temperatures but liquid at the rectal temperature and will
therefore melt in the
rectum and release the drug.
The pharmaceutical compositions may be subjected to conventional
pharmaceutical
operations such as sterilization and/or may contain conventional adjuvants,
such as
preservatives, stabilizers, wetting agents, emulsifiers, buffers etc. Tablets
and pills can
additionally be prepared with enteric coatings. Such compositions may also
comprise
adjuvants, such as wetting, sweetening, flavoring, and perfuming agents.
The foregoing is merely illustrative of the invention and is not intended to
limit the
invention to the disclosed compounds. Variations and changes, which are
obvious to one
skilled in the art are intended to be within the scope and nature of the
invention, which are
defined, in the appended claims.



CA 02703257 2012-03-13

WO 2009/058267 PCT/1JS2008/012224
From the foregoing description, one skilled in the art can easily ascertain
the
characteristics of this invention. The claims are to be given a purposive
construction
based on the application as a whole.
No unacceptable toxological effects are expected when compounds of the
present invention are administered in accordance with the present invention.

56

Representative Drawing

Sorry, the representative drawing for patent document number 2703257 was not found.

Administrative Status

For a clearer understanding of the status of the application/patent presented on this page, the site Disclaimer , as well as the definitions for Patent , Administrative Status , Maintenance Fee  and Payment History  should be consulted.

Administrative Status

Title Date
Forecasted Issue Date 2013-02-19
(86) PCT Filing Date 2008-10-28
(87) PCT Publication Date 2009-05-07
(85) National Entry 2010-04-20
Examination Requested 2010-04-20
(45) Issued 2013-02-19
Deemed Expired 2018-10-29

Abandonment History

There is no abandonment history.

Payment History

Fee Type Anniversary Year Due Date Amount Paid Paid Date
Request for Examination $800.00 2010-04-20
Application Fee $400.00 2010-04-20
Maintenance Fee - Application - New Act 2 2010-10-28 $100.00 2010-09-17
Maintenance Fee - Application - New Act 3 2011-10-28 $100.00 2011-09-20
Maintenance Fee - Application - New Act 4 2012-10-29 $100.00 2012-09-19
Final Fee $300.00 2012-12-07
Maintenance Fee - Patent - New Act 5 2013-10-28 $200.00 2013-09-13
Maintenance Fee - Patent - New Act 6 2014-10-28 $200.00 2014-10-08
Maintenance Fee - Patent - New Act 7 2015-10-28 $200.00 2015-10-07
Maintenance Fee - Patent - New Act 8 2016-10-28 $200.00 2016-10-05
Owners on Record

Note: Records showing the ownership history in alphabetical order.

Current Owners on Record
AMGEN INC.
Past Owners on Record
DILMEGHANI SERAN, MINA
HARMANGE, JEAN-CHRISTOPHE
LARROW, JAY
MARTIN, MATTHEW W.
PAYACK, JOSEPH F.
SUBRAMANIAN, RAJU
TEFFERA, YOHANNES
WHITE, RYAN
ZANON, ROGER
Past Owners that do not appear in the "Owners on Record" listing will appear in other documentation within the application.
Documents

To view selected files, please enter reCAPTCHA code :



To view images, click a link in the Document Description column. To download the documents, select one or more checkboxes in the first column and then click the "Download Selected in PDF format (Zip Archive)" or the "Download Selected as Single PDF" button.

List of published and non-published patent-specific documents on the CPD .

If you have any difficulty accessing content, you can call the Client Service Centre at 1-866-997-1936 or send them an e-mail at CIPO Client Service Centre.


Document
Description 
Date
(yyyy-mm-dd) 
Number of pages   Size of Image (KB) 
Abstract 2010-04-20 1 64
Claims 2010-04-20 5 92
Description 2010-04-20 56 2,658
Claims 2010-04-21 5 97
Cover Page 2010-06-18 2 37
Description 2012-03-13 56 2,636
Claims 2012-03-13 3 45
Claims 2012-06-08 3 48
Cover Page 2013-01-24 2 37
PCT 2010-04-20 1 44
Assignment 2010-04-20 6 160
Prosecution-Amendment 2010-04-20 2 47
PCT 2010-09-03 1 50
Prosecution-Amendment 2011-11-25 2 89
Prosecution-Amendment 2012-03-13 12 427
Correspondence 2012-06-08 3 78
Correspondence 2012-06-04 1 22
Correspondence 2012-12-07 2 50